EP2605791B1 - Préparation d'immunoglobuline enrichie en anti-lps, destinée à être utilisée dans le traitement et/ou la prophylaxie d'un trouble pathologique - Google Patents

Préparation d'immunoglobuline enrichie en anti-lps, destinée à être utilisée dans le traitement et/ou la prophylaxie d'un trouble pathologique Download PDF

Info

Publication number
EP2605791B1
EP2605791B1 EP11793860.5A EP11793860A EP2605791B1 EP 2605791 B1 EP2605791 B1 EP 2605791B1 EP 11793860 A EP11793860 A EP 11793860A EP 2605791 B1 EP2605791 B1 EP 2605791B1
Authority
EP
European Patent Office
Prior art keywords
cells
lps
composition
immunoglobulin preparation
colostrum
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Not-in-force
Application number
EP11793860.5A
Other languages
German (de)
English (en)
Other versions
EP2605791A2 (fr
Inventor
Tomer Adar
Meir Mizrahi
Gadi Lalazar
Yaron Ilan
Ami Ben-Ya'acov
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Immuron Ltd
Original Assignee
Immuron Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Immuron Ltd filed Critical Immuron Ltd
Publication of EP2605791A2 publication Critical patent/EP2605791A2/fr
Application granted granted Critical
Publication of EP2605791B1 publication Critical patent/EP2605791B1/fr
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1203Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/40Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum bacterial
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/12Immunoglobulins specific features characterized by their source of isolation or production isolated from milk

Definitions

  • the invention relates preparations enriched with anti LPS antibodies for use in the treatment of non-alcoholic steatohepatitis (NASH), such as those derived from mammalian colostrum,
  • NASH non-alcoholic steatohepatitis
  • Chronic hepatitis is inflammation of the liver that lasts at least six months. Chronic hepatitis, although much less common than acute hepatitis, can persist for years, even decades. In most people, it is quite mild and does not cause significant liver damage. However, in some people, continued inflammation slowly damages the liver, eventually resulting in cirrhosis (severe scarring of the liver), liver failure, and sometimes liver cancer. Chronic hepatitis is usually caused by Hepatitis B virus (HBV) Hepatitis C virus (HCV) and drugs.
  • HBV Hepatitis B virus
  • HCV Hepatitis C virus
  • Chronic Hepatitis C virus (HCV) infection is characterized by the inability of the host to establish an effective immune response.
  • chronic HCV infection is associated with persistent, abnormally high levels of immune activation. This activation contributes to liver damage and disease progression.
  • Patients with chronic HCV infection have widely varying clinical courses, while some develop cirrhosis, others do not show progression of liver disease.
  • LPS may not only be connected with progression of liver disease, but also with the perpetuation of chronic infection.
  • monocytes from patients with chronic HCV produce significantly more IL-10 and TNF alpha in response to the HCV core protein or LPS ( Martin-Blondel, G. et al. J. Viral. Hepatitis (March 11, 2009 )).
  • LPS LPS core protein
  • Spontaneous bacterial peritonitis is a common and severe complication of chronic liver diseases, such as liver cirrhosis, portal hypertension and ascites. SBP occurs in up to 30% of patients, and is associated with an in-hospital mortality rate of up to 25%. Bacterial translocation into the stagnant and immune depleted peritoneal fluid is considered to be the main pathogenic mechanism of SBP. While paracentesis and broad spectrum antibiotic therapy constitute an effective treatment for acute infection, many patients suffer from recurrent episodes of SBP with pathogens which become increasingly resistant to antibiotic therapy ( Song, K.H. et al. BMC Infect Dis. 9(1): 41 (2009 )). Methods for SBP prophylaxis using chronic antibiotics are controversial and associated with immergence of antibiotic resistant species ( Cohen, M.J. et al. Cochrane Database Syst Rev. 2: CD004791 (2009 )).
  • Colostrum also known as first milk
  • first milk is a form of milk produced by the mammary glands in late pregnancy and the few days after birth. In humans it has high concentrations of nutrients and antibodies, but it is small in quantity. Colostrum is high in carbohydrates, protein, mineral salts, vitamins and immunoglobulin. It also contains various floating cells such as granular and stromal cells, neutrophils, monocyte/macrophages and lymphocytes and includes growth factors, hormones and cytokines.
  • Leukocytes are also present in colostrum in large numbers which enable protection against viruses and bacteria.
  • Colostral leukocytes enhance passive immunity of neonatal calf, especially in regard to antibodies and immunoglobulin classes which are essential for intestinal immunity.
  • Bovine colostrum contains three major classes of immunoglobulins: IgG, IgM and IgA.
  • colostrum is quite a unique product that arises from a distinct physiological and functional state of the mammary gland.
  • the principal compositional difference between colostrum and mature milk is the very high content of bioactive components such as lactoferrin and immunoglobulins ( Tarbell, K.V. et al. J. Exp. Med. 199:1467-77 (2004 ); Bluestone, J.A. and Tang, Q. J. Autoimmun 24:55-62 (2005 ); Putnam, A.l. et al J.Autoimmun. 24:55-62 (2005 )), of which IgG class makes up 80-90%.
  • the immunization of an animal such as a cow with specific antigens enables the production and harvest of specific antibodies that may be used for modulation of an immune response and thereby in the treatment of immune-related disorders. Accordingly, this method serves as an easy and safe means for generating antigen-specific antibodies and immune adjuvants.
  • WO 04/078209 by some of the present inventors describes compounds and compositions for the treatment or prophylaxis of gastrointestinal disorders prepared by immunizing a host animal with a vaccine comprising one or more cell wall antigens of enteric bacteria, specifically, gram negative bacteria.
  • the hyper immune material produced is in the form of tablets for oral administration.
  • WO 03/097094 describes the use of a hyper immune colostrum in the production of antibodies (whole IgG), or F(ab')2 antibodies fragments, conjugated with mammalian colostrum and colostrum extracts, for intranasal administration aimed at the prevention of symptoms arising from the presence of air-borne pathogenic bacteria.
  • Mucosal tolerance is considered as an attractive approach for the treatment of autoimmune and inflammatory diseases due to the lack of toxicity, ease of administration, and antigen-specific mechanism of action ( Wershil, B.K. and Furuta, G.T. J. Allergy Clin. Immunol. 121:S380-3 ; quiz S415 (2008); Faria, A.M. and Weiner, H.L. Clin. Dev. Immunol. 13:143-57 (2006 )).
  • major attempts were made to generate stable colostrum-derived products suitable for oral and nasal administration.
  • WO 95/08562 describes the method of obtaining high purity immunoglobulins from antibody rich colostrum and the possibility of compressing these colostral-antibodies into a tablet form without substantial loss of activity.
  • Specific antibodies may be obtained by immunization of a mammal with specific antigens against enterotoxic bacteria such as E. coli, Salmonela and Shigella.
  • WO 06/053383 describes a carboxylic acid and alkalizing moieties which confer upon a bioactive agent composition of a hyper immune colostrum, lactoferrin or lactoferracin, stability under a wide variety of gastric pH values.
  • WO 03/080082 by some of the inventors describes a method of improving the viability of a labile bioactive substance, preferably immunoglobulins or fragments thereof or enzymes, in a gastric environment, comprising forming a mixture of the bioactive substance and mammalian colostrum and colostrums extracts.
  • This conjugation protects the antibodies or antibodies fragments from the proteolysis occasioned by enzyme or low pH conditions and preserves their function in the stomach or rumen or other hostile environment.
  • the bowel mucosa is the largest lymphoid organ of the body. It deals with the dual role of nutrient absorption, while maintaining a physical and immunological barrier to the gut content. Despite constant antigenic stimulation, suppression of inflammation is the rule.
  • Two key concepts pertain to the treatment of viral disease and its complications with colostrums: mucosal microbial translocation and enhanced immune regulation by oral feeding of disease antigens, termed "oral tolerance”.
  • Increased mucosal microbial translocation is an immerging concept in disease pathogenesis.
  • the higher levels of microbial translocation, quantified by the presence of LPS and bacterial DNA are central to a state of chronic immune activation accounting for immune exhaustion and autoimmune damage.
  • Stimulation through the bowel mucosa tends to elicit a tolerogenic immune response.
  • This feature may be used advantageously to induce tolerance towards auto-antigens and in this way to suppress autoimmunity.
  • oral tolerance has been shown to effectively diminish the immune response towards orally fed antigens in different disease models ( Safadi, R. et al. Am. J. Gastroenterol. 98(11): 2505-15 (2003 )).
  • bovine-derived colostrum preparations can be used in treating toxin-mediated intestinal conditions.
  • administration of a bovine antibody concentrate obtained by immunizing cows with the corresponding E. coli strains prevented the development of diarrhea in all 10 participants who received the product; by contrast, 9/10 controls developed diarrhea ( Tacket, C.O. et al. N. Engl. J. Med. 318(19): 1240-3 (1988 )).
  • the administration of milk-derived antibodies against the enterotoxigenic E. coli colonization factor protected 14/15 subjects from diarrhea, compared to 7/10 subjects given placebo ( Freedman, D.J. et al.
  • WO2004/078209 discloses the treatment of gastrointestinal disorders by administering a vaccine ir hyper immune material that is rasied agains lipopolysaccharide (LPS) associated antigens.
  • Another disease with a similar pathogenesis is pseudomembranous colitis.
  • WO2009/113065 discloses immunomodulatory compositions comprising mammalian colostrum-derived immunoglobulin preparations for treating immune-related disorders.
  • bovine-derived colostrum preparations deliver biologically active concentrations of specific antibodies to the intestinal lumen when taken orally, and might be capable of blocking various forms of bacterial toxins in the gut by that mechanism.
  • bovine colostrum powder (BPC) preparations from immunized cows, containing high levels of antibodies, as immuno-modulators capable of reducing immune activation in response to microbial products such as LPS.
  • BPC bovine colostrum powder
  • the inventors hypothesize that the attachment of the BPC antibodies to the microbial antigens may prevent their translocation onto the blood stream, thereby restricting the immune response.
  • Microbial translocation is also associated with alteration of the liver inflammation in different liver disorders, including viral mediated, drug mediated, non-alcoholic steatohepatitis and any other hepatic disorder. Microbial translocation may also be associated with insulin resistance, diabetes type 2, obesity and overweight. As shown by the disclosure, prevention of such translocation may be achieved using the anti-LPS enriched colostrum of the disclosure, optionally along with regulation of regulatory T cells, or any other component of the immune system, using a combination of the anti-LPS enriched colostrum with colostrum preparations enriched with antibodies recognizing disease-specific antigens, for example, anti-insulin enriched colostrum. Thus, the disclosure further provides compositions, combined compositions and methods for the treatment of any acute or chronic liver disease, diabetes and any complication associated therewith, fatty liver, non-alcoholic steatohepatitis, and obesity.
  • Another object of the invention is to provide combined compositions comprising a combination of anti-LPS antibodies enriched colostrum and antibodies recognizing at least one antigen specific for a pathologic disorder and uses thereof in the treatment of immune-related disorders.
  • the present invention is based, at least in part, on the discovery that subjects who suffer from non-alcoholic steatohepatitis (NASH) can be treated using an anti-LPS immunoglobulin-enriched colostrum preparation.
  • NASH non-alcoholic steatohepatitis
  • compositions for use in treating such subjectsin accordance with the claims are provided herein.
  • the invention provides compositions comprising an anti-LPS immunoglobulin-enriched colostrum preparation, e.g., a bovine colostrum preparation, as described herein for use the treatment of non-alcoholic steatohepatitis (NASH) in a subject who has one or more of: an ALT level above 50 IU/dL, an AST level above 50 IU/dL, an AP of greater than 70 U/L, and/or a GGT of greater than 60 U/L, wherein the composition is formulated for oral administration, and the oral dose form comprises 200 mg to 2000 mg IgG with the anti-LPS immunoglobulin being at least 7% dry weight of the composition of IgG.
  • NASH non-alcoholic steatohepatitis
  • the disclosure provides methods for selecting a treatment to improve liver function in a subject.
  • the methods include detecting the presence in a sample from the subject of one or more of: an ALT level above 50 IU/dL, an AST level above 50 IU/dL, an AP of greater than 70 U/L, and/or a GGT of greater than 60 U/L; and selecting a treatment comprising administration of a composition comprising an anti-LPS immunoglobulin-enriched colostrum preparation for the subject.
  • the methods further include administering the treatment to the subject.
  • the dislcosure provides compositions including an anti-LPS immunoglobulin-enriched colostrum preparation as described herein for use in improving insulin resistance in a subject who has one or more of a level of HbAlc above 7, and/or a level of insulin secretion above 300 pg/ml.
  • the disclosure provides methods for selecting a treatment to improve insulin resistance in a subject.
  • the methods include detecting the presence in a sample from the subject of one or more of: a level of HbA1c above 7, and/or a level of insulin secretion above 300 pg/ml, and selecting a treatment comprising administration of a composition comprising an anti-LPS immunoglobulin-enriched colostrum preparation for the subject.
  • the methods further include administering the treatment to the subject.
  • the subject has metabolic syndrome or diabetes.
  • the composition reduces serum insulin levels, increases serum insulin levels, improves fasting glucose levels, reduces HbAlc levels, or both.
  • the disclosure features methods for selecting a treatment to improve serum cholesterol levels in a subject.
  • the methods include detecting the presence in a sample from the subject of one or more of: a level of total serum cholesterol above the upper limit of normal; a level of serum LDL above the upper limit of normal; a level of serum triglycerides above the upper limit of normal; and selecting a treatment comprising administration of a composition comprising an anti-LPS immunoglobulin-enriched colostrum preparation for the subject.
  • the methods further include administering the treatment to the subject.
  • compositions including an anti-LPS immunoglobulin enriched colostrum preparation as described herein for use in increasing levels of GLP-1 or adiponectin in a subject.
  • compositions including an anti-LPS immunoglobulin enriched colostrum preparation as described herein for use in treating sepsis in a subject.
  • compositions including an anti-LPS immunoglobulin enriched colostrum preparation as described herein for use in treating inflammatory bowel disease in a subject.
  • the anti-LPS immunoglobulin preparation is prepared by immunizing a cow with LPS comprising 06, 08, 015, 025, 027, 063, 078, 0114, 0115, 0128, 0148, 0153, and 0159.
  • the anti-LPS immunoglobulin preparation is administered at a dose of about 200 mg to about 2000 mg per day, e.g., at a dose of about 1800 mg per day.
  • an anti-LPS enriched immunoglobulin preparation e.g., an anti-LPS immunoglobulin-enriched colostrum preparation, e.g., a bovine colostrum preparation, for use in treatment and/or prophylaxis of a pathologic disorder.
  • the anti-LPS enriched immunoglobulin preparation may be derived from colostrum or from avian eggs.
  • the anti-LPS enriched immunoglobulin preparation is an anti-LPS immunoglobulin-enriched colostrum preparation, e.g., a bovine colostrum preparation, produced by a method described herein.
  • the pathologic disorder is acute or chronic liver disease, cirrhosis or any disease or complication associated therewith.
  • the acute or chronic liver disease, cirrhosis and any disease or complication associated therewith is selected from the group consisting of hepatic encephalopathy, spontaneous bacterial peritonitis (SBP), ascites, bleeding varices, cirrhosis associated hyperdynamic circulation, hepatorenal syndrome, hepatopulmonary syndrome, portopulmonary hypertension, variceal bleeding, adrenal insufficiency and altered level of consciousness.
  • the pathologic disorder is liver damage.
  • the pathologic disorder is an immune-related disorder selected from the group consisting of autoimmune disease, non-alcoholic steatohepatitis (NASH), fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith, infectious disease, and proliferative disorder.
  • autoimmune disease non-alcoholic steatohepatitis (NASH)
  • NASH non-alcoholic steatohepatitis
  • fatty liver fatty liver
  • atherosclerosis fatty liver
  • metabolic syndrome any disorder associated therewith
  • infectious disease infectious disease
  • proliferative disorder proliferative disorder
  • the pathologic disorder may be selected from the group consisting of secondary peritonitis and infection after surgery, hepatic cardiomyopathy and hypotension, hepatoadrenal syndrome, hepatocellular carcinoma, Alzheimer's disease, any type of memory loss, any type of dementia, attention deficit disorders (ADHA), any type of learning disability, effect of alcohol or drugs on the brain, any type of immune mediated disease including asthma, and peritonitis.
  • the composition further comprises an immunoglobulin preparation comprising immunoglobulins that recognize and bind at least one antigen specific for said pathologic disorder.
  • the further immunoglobulin preparation may be derived from colostrum.or from avian eggs.
  • the composition modulates regulatory T cells leading to modulation of the Thl/Th2, Trl/Th3 cell balance toward an anti-inflammatory Th2, Trl/Th3 immune response or a pro-inflammatory Th1 immune response thereby inhibiting or activating an immune response specifically directed toward said disorder.
  • the composition modulates the Th1/Th2, Trl/Th3 cell balance toward an anti-inflammatory Th2, Trl/Th3 immune response thereby inhibiting an immune response specifically directed toward said disorder, and wherein said composition is for the treatment of any one of an autoimmune disease, non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith selected from diabetes type 2, insulin resistance, obesity and overweight.
  • the composition is for the treatment and/or prophylaxis of metabolic syndrome or non-alcoholic steatohepatitis or both.
  • the composition is for the treatment, and/or prophylaxis of diabetes, the treatment of impaired glucose tolerance, such as decreasing glucose tolerance. decreasing serum insulin levels, decreasing hepatic triglyceride levels, or decreasing cholesterol levels.
  • the composition modulates the Th1/Th2, Trl/Th3 cell balance toward a pro-inflammatory Th1/Th2 immune response thereby enhancing an immune response specifically directed toward said disorder, and wherein said composition is for the treatment of infectious diseases, and proliferative disorders.
  • composition may further comprise a therapeutic agent, carrier or adjuvant and/or non-hyperimmune colostrum.
  • composition may be formulated for administration orally, by inhalation as an aerosol, or by parenteral, intravaginal, intranasal, mucosal, sublingual, topical, or rectal administration, or any combination thereof.
  • the colostrum preparation or any fractions thereof comprise immunoglobulins that recognize and bind LPS.
  • the composition inhibits microbial translocation. In some embodiments the composition inhibits microbial translocation and thereby modulates immune activation.
  • the present disclosure provides a composition comprising a mammalian anti-LPS enriched colostrum-derived immunoglobulin preparation for modulating immune tolerance in a subject, or in another aspect, for modulating oral tolerance in a subject.
  • the present disclosure provides a composition comprising a mammalian anti-LPS immunoglobulin enriched colostrum-derived preparation for inducing CD4+ CD25+ T cells in the liver, inducing CD4+ CD25+ LAP- T cells in the liver, inducing CD45+ LAP+ T cells in the liver, inducing CD3+ LAP+ T cells in the liver, inducing CD45+ LAP+ T cells in the spleen, inducing CD8+ LAP+ T cells in the spleen, inducing CD3+ LAP+ T cells in the spleen, inducing CD8+ CD25+ T cells in the spleen, inducing CD4+ CD25+ T cells in adipose tissue, inducing CD3+ LAP+ T cells in adipose tissue, inducing CD4+ CD25+ T cells in stromal vascular cells, inducing CD4+ CD25+ T cells in stromal vascular cells, decreasing CD
  • the anti-LPS enriched immunoglobulin preparation described as part of the disclosure herein may be derived from colostrum or from avian eggs.
  • the preparation is an anti-LPS immunoglobulin-enriched colostrum preparation, e.g., a bovine colostrum preparation, produced by a method described herein.
  • the present disclosure provides a use of an anti-LPS enriched immunoglobulin preparation in the manufacture of a medicament for the treatment and/or prophylaxis of a pathologic disorder.
  • the anti-LPS enriched immunoglobulin preparation of the disclosure may be derived from colostrum or from avian eggs.
  • the pathologic disorder is acute or chronic liver disease, cirrhosis or any disease or complication associated therewith.
  • the acute or chronic liver disease, cirrhosis and any disease or complication associated therewith is selected from the group consisting of hepatic encephalopathy, spontaneous bacterial peritonitis (SBP), ascites, bleeding varices, cirrhosis associated hyperdynamic circulation, hepatorenal syndrome, hepatopulmonary syndrome, portopulmonary hypertension, variceal bleeding, adrenal insufficiency and altered level of consciousness.
  • SBP spontaneous bacterial peritonitis
  • the medicament is for the treatment and/or prophylaxis of liver damage.
  • the pathologic disorder is an immune-related disorder selected from the group consisting of autoimmune disease, non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith, infectious disease, and proliferative disorder.
  • the pathologic disorder is selected from the group consisting of secondary peritonitis and infection after surgery, hepatic cardiomyopathy and hypotension, hepatoadrenal syndrome, hepatocellular carcinoma, Alzheimer's disease, any type of memory loss, any type of dementia, attention deficit disorders (ADHA), any type of learning disability, effect of alcohol or drugs on the brain, any type of immune mediated disease including asthma, and peritonitis.
  • the medicament may further comprise an immunoglobulin preparation comprising immunoglobulins that recognize and bind at least one antigen specific for said pathologic disorder.
  • the further immunoglobulin preparation may be derived from colostrum.or from avian eggs.
  • the medicament modulates regulatory T cells leading to modulation of the Th1/Th2, Trl/Th3 cell balance toward an anti-inflammatory Th2, Trl/Th3 immune response or a pro-inflammatory Th1 immune response thereby inhibiting or activating an immune response specifically directed toward said disorder.
  • the medicament modulates the Th1/Th2, Trl/Th3 cell balance toward an anti-inflammatory Th2, Trl/Th3 immune response thereby inhibiting an immune response specifically directed toward said disorder
  • said composition is for the treatment of any one of an autoimmune disease, non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith selected from diabetes type 2, insulin resistance, obesity and overweight.
  • the medicament is for the treatment and/or prophylaxis of metabolic syndrome or non-alcoholic steatohepatitis or both, the treatment and/or prophylaxis of diabetes, the treatment impaired glucose tolerance, such as decreasing glucose tolerance, decreasing serum insulin levels, decreasing hepatic triglyceride levels, or decreasing cholesterol levels.
  • the medicament modulates the Th1/Th2, Trl/Th3 cell balance toward a pro-inflammatory Th1/Th2 immune response thereby enhancing an immune response specifically directed toward said disorder, and wherein said composition is for the treatment of infectious diseases, and proliferative disorders,
  • the medicament may further comprise a therapeutic agent, carrier or adjuvant and/or non-hyperimmune colostrum.
  • the medicament is formulated for administration orally, by inhalation as an aerosol, or by parenteral, intravaginal, intranasal, mucosal, sublingual, topical, or rectal administration, or any combination thereof.
  • the immunoglobulin preparation or any fractions thereof recognizes and binds LPS or any fragments thereof.
  • the composition reduces or inhibits mucosal microbial translocation and thereby modulates immune activation.
  • the present disclosure provided a use of a mammalian anti-LPS enriched colostrum-derived immunoglobulin preparation in the manufacture of a medicament for modulating immune tolerance in a subject, or in another embodiment, a medicament for modulating oral tolerance in a subject.
  • the present disclosure provides the use of a mammalian anti-LPS enriched colostrum-derived immunoglobulin preparation in the manufacture of a medicament for inducing CD4+ CD25+ T cells in the liver, inducing CD4+ CD25+ LAP- T cells in the liver, inducing CD45+ LAP+ T cells in the liver, inducing CD3+ LAP+ T cells in the liver, inducing CD45+ LAP+ T cells in the spleen, inducing CD8+ LAP+ T cells in the spleen, inducing CD3+ LAP+ T cells in the spleen, inducing CD8+ CD25+ T cells in the spleen, inducing CD4+ CD25+ T cells in adipose tissue, inducing CD3+ LAP+ T cells in adipose tissue, inducing CD4+ CD25+ T cells in stromal vascular cells, inducing CD4+ CD25+ LAP+ T cells in stromal
  • the anti-LPS enriched immunoglobulin preparation of the disclosure may be derived from colostrum or from avian eggs.
  • the present disclosure provides a method for the treatment and/or prophylaxis of a pathologic disorder comprising the step of administering to a subject in need thereof a therapeutically effective amount of a composition comprising an anti-LPS enriched immunoglobulin preparation.
  • the anti-LPS enriched immunoglobulin preparation may be derived from colostrum or from avian eggs.
  • the pathologic disorder is acute or chronic liver disease, cirrhosis or any disease or complication associated therewith.
  • the acute or chronic liver disease, cirrhosis and any disease or complication associated therewith is selected from the group consisting of hepatic encephalopathy, spontaneous bacterial peritonitis (SBP), ascites, bleeding varices, cirrhosis associated hyperdynamic circulation, hepatorenal syndrome, hepatopulmonary syndrome, portopulmonary hypertension, variceal bleeding, adrenal insufficiency and altered level of consciousness.
  • SBP spontaneous bacterial peritonitis
  • the pathologic disorder is liver damage.
  • the pathologic disorder is an immune-related disorder selected from the group consisting of autoimmune disease, non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith, infectious disease, and proliferative disorder.
  • the pathologic disorder is selected from the group consisting of secondary peritonitis and infection after surgery, hepatic cardiomyopathy and hypotension, hepatoadrenal syndrome, hepatocellular carcinoma, Alzheimer's disease, any type of memory loss, any type of dementia, attention deficit disorders (ADHA), any type of learning disability, effect of alcohol or drugs on the brain, any type of immune mediated disease including asthma, and peritonitis.
  • the composition further comprises an immunoglobulin preparation comprising immunoglobulins that recognize and bind at least one antigen specific for said pathologic disorder.
  • the further immunoglobulin preparation may be derived from colostrum.or from avian eggs.
  • the composition modulates regulatory T cells leading to modulation of the Th1/Th2, Trl/Th3 cell balance toward an anti-inflammatory Th2, Trl/Th3 immune response or a pro-inflammatory Th1 immune response thereby inhibiting or activating an immune response specifically directed toward said disorder.
  • the composition modulates the Th1/Th2, Trl/Th3 cell balance toward an anti-inflammatory Th2, Trl/Th3 immune response thereby inhibiting an immune response specifically directed toward said disorder, and wherein said composition is for the treatment of any one of an autoimmune disease, non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith selected from diabetes type 2, insulin resistance, obesity and overweight.
  • the pathologic disorder is metabolic syndrome or non-alcoholic steatohepatitis or both.
  • the pathologic disorder is diabetes. In another embodiment, the pathologic disorder is impaired glucose tolerance.
  • the method decreases glucose tolerance, decreases serum insulin levels, decreases hepatic triglyceride levels, or decreases cholesterol levels.
  • the method modulates the Th1/Th2, Trl/Th3 cell balance toward a pro-inflammatory Th1/Th2 immune response thereby enhancing an immune response specifically directed toward said disorder, and wherein said composition is for the treatment of infectious diseases, and proliferative disorders,
  • composition further comprises non-hyperimmune colostrum and/or a therapeutic agent, carrier or adjuvant.
  • composition may be administered orally, by inhalation as an aerosol, or by parenteral, intravaginal, intranasal, mucosal, sublingual, topical, or rectal administration, or any combination thereof.
  • the immunoglobulin preparation or any fractions thereof recognizes and binds LPS or any fragments thereof.
  • the method reduces or inhibits mucosal microbial translocation. In another embodiment, the method reduces or inhibits mucosal microbial translocation and thereby modulates immune activation.
  • the present disclosure provides a method for modulating immune tolerance in a subject comprising the step of administering to a subject in need thereof a therapeutically effective amount of a composition comprising a mammalian anti-LPS enriched colostrum-derived immunoglobulin preparation.
  • the method may be for modulating oral tolerance.
  • a method for inducing CD4+ CD25+ T cells in the liver of a subject comprising the step of administering to a subject in need thereof a therapeutically effective amount of a composition comprising a mammalian anti-LPS enriched colostrum-derived immunoglobulin preparation.
  • the method may be for inducing CD4+ CD25+ LAP- T cells in the liver, CD45+ LAP+ T cells in the liver, inducing CD3+ LAP+ T cells in the liver, inducing CD45+ LAP+ T cells in the spleen, inducing CD8+ LAP+ T cells in the spleen, inducing CD3+ LAP+ T cells in the spleen, inducing CD8+ CD25+ T cells in the spleen, inducing CD4+ CD25+ T cells in adipose tissue, inducing CD3+ LAP+ T cells in adipose tissue, inducing CD4+ CD25+ T cells in stromal vascular cells, inducing CD4+ CD25+ LAP+ T cells in stromal vascular cells, decreasing CD3+ NK1.1+ cells in the liver, decreasing CD25+ LAP- T cells in the liver, decreasing CD25+ LAP+ T cells in the liver, inducing CD4+ CD25+
  • the present disclosure provides a composition for the treatment and prophylaxis of a pathologic disorder.
  • the composition of the invention comprises as active ingredient a mammalian anti-lipopolysaccharide (anti-LPS) enriched colostrum-derived immunoglobulin preparation and optionally further colostrum, milk or milk product component/s, and any adjuvant/s.
  • the immunoglobulin preparation or any fractions thereof recognizes and binds LPS and any fragments thereof.
  • the composition of the invention may further comprises colostrum-derived immunoglobulin preparation recognizing at least one antigen specific for said disorder, thereby activating or inhibiting an immune response specifically directed toward said disorder.
  • Such combined composition may optionally further comprises an additional therapeutic agent or any carrier and adjuvant.
  • the disclosure provides a composition
  • a composition comprising as an active ingredient a mammalian anti-lipopolysaccharide (LPS) enriched colostrum-derived immunoglobulin preparation.
  • LPS mammalian anti-lipopolysaccharide
  • Such composition wherein said composition is particularly applicable for the treatment, prevention and prophylaxis of acute or chronic liver disease, cirrhosis and any disease or complication associated therewith, optionally said composition further comprises an additional therapeutic agent or any carrier and adjuvant.
  • the disclosure provides combined compositions comprising a combination of anti-LPS enriched immunoglobulin preparation with at least one colostrum- or avian- derived immunoglobulin preparation comprising immunoglobulins that recognize and bind at least one antigen specific for said pathologic disorder.
  • Such combined composition may optionally further comprises an additional therapeutic agent or any carrier and adjuvant.
  • These combined compositions may be used for treating any one of an autoimmune disease, non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith such as diabetes type 2, insulin resistance, obesity and overweight.
  • the present disclosure provides the use of a mammalian anti-LPS enriched colostrum-derived immunoglobulin preparation and optionally of a colostrum-derived immunoglobulin preparation recognizing at least one antigen specific for a pathologic disorder in the manufacture of a composition for the treatment and prophylaxis of a pathologic disorder, It should be noted that the immunoglobulin preparation or any fractions thereof recognizes and binds LPS and any fragments thereof. According to an optional embodiment, the disclosure provides the use of the anti-LPS enriched immunoglobulin preparation of the disclosure furthering combination with at least one immunoglobulin preparation comprising immunoglobulins recognizing at least one antigen specific for said disorder. Such combined composition may be used as an immuno-modulatory composition that activates or inhibits an immune response specifically directed toward said disorder.
  • the present disclosure provides a method for the treatment and/or prophylaxis of a pathologic disorder.
  • the method of the disclosure comprises the step of administering to a subject in need thereof a therapeutically effective amount of a mammalian colostrum-derived anti-LPS enriched immunoglobulin preparation or of a composition comprising the same.
  • a mammalian colostrum-derived anti-LPS enriched immunoglobulin preparation or of a composition comprising the same a mammalian colostrum-derived anti-LPS enriched immunoglobulin preparation or of a composition comprising the same.
  • the immunoglobulin preparation or any fractions thereof recognizes and binds LPS and any fragments thereof.
  • Such method may be used for the treatment, prevention and prophylaxis of acute or chronic liver disease, cirrhosis and any disease or complication associated therewith.
  • the anti-LPS enriched immunoglobulin preparation of the disclosure may be further combined with at least one immunoglobulins recognizing at least one antigen specific for said disorder, thereby activating or inhibiting an immune response specifically directed toward said disorder.
  • This method may be specifically applicable for treating immune-related disorders. It should be particularly appreciated that the compositions and combined compositions used by the methods of the disclosure may be also applicable for treating any one of non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith such as diabetes type 2, insulin resistance, obesity and overweight.
  • the present disclosure provides a method for treating a human subject with a condition selected from the group consisting of hypertension, increase in body mass index (BMI), increase in waist circumference, dyslipidemia , insulin resistance, elevated liver enzymes, and fatty liver comprising administering to the subject an effective amount of a composition comprising an anti-LPS immunoglobulin preparation.
  • a condition selected from the group consisting of hypertension, increase in body mass index (BMI), increase in waist circumference, dyslipidemia , insulin resistance, elevated liver enzymes, and fatty liver comprising administering to the subject an effective amount of a composition comprising an anti-LPS immunoglobulin preparation.
  • compositions comprising an anti-LPS immunoglobulin preparation for use in treatment and/or prophylaxis of a human subject with a condition selected from the group consisting of hypertension, increase in body mass index (BMI), increase in waist circumference, dyslipidemia , insulin resistance, elevated liver enzymes, and fatty liver.
  • BMI body mass index
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for the treatment and/or prophylaxis of a human subject with a condition selected from the group consisting of hypertension, increase in body mass index (BMI), increase in waist circumference, dyslipidemia , insulin resistance, elevated liver enzymes, and fatty liver.
  • a condition selected from the group consisting of hypertension, increase in body mass index (BMI), increase in waist circumference, dyslipidemia , insulin resistance, elevated liver enzymes, and fatty liver.
  • the hypertension may characterized by a blood pressure of >120 mmHg / 80 mmHg, a blood pressure of >130 mmHg / 90 mmHg or a blood pressure of >140 mmHg / 90 mmHg.
  • the fatty liver may be characterized by macrovesicular steatosis, macrovesicular steatosis and necroinflammatory activity, or a NAS score of at least 4.
  • the anti-LPS immunoglobulin preparation may be derived from colostrum or avian eggs.
  • the anti-LPS immunoglobulin preparation may be administered at a dose of about 5 mg to about 25000 mg per day, 10 mg to about 20000 mg per day, 25 mg to about 15000 mg per day, 100 mg to about 2000 mg per day, or about 1800 mg per day. In one embodiment, the anti-LPS immunoglobulin preparation is not administered at a dose of about 600 mg per day.
  • the anti-LPS immunoglobulin preparation may be formulated for administration at a dose of about 5 mg to about 25000 mg per day, about 10 mg to about 20000 mg per day, about 25 mg to about 15000 mg per day, about 100 mg to about 2000 mg per day or about 1800 mg per day. In one embodiment, the anti-LPS immunoglobulin preparation is not formulated for administration at a dose of about 600 mg per day
  • the anti-LPS immunoglobulin preparation may be prepared by immunizing a mammal or avian with LPS from multiple E.coli strains.
  • the mammal or avian may be immunized with LPS selected from the group consisting of 06, 08, 015, 025, 027, 063, 078, 0114, 0115, 0128, 0148, 0153, 0159, and other LPS associated with enterotoxigenic E. coli.
  • the mammal or avian may be immunized with LPS selected from the group consisting of O78, O6, O8, O129 and 0153 LPS.
  • LPS may comprise 078 LPS.
  • composition further comprises an anti-insulin immunoglobulin preparation.
  • the anti-insulin immunoglobulin preparation is administered at a dose of about 5mg to about 25000mg per day, about 10 mg to about 20000 mg per day, about 25 mg to about 15000 mg per day, about 50 mg to about 10000 mg per day, about 50 mg to about 4000mg per day, about 500 mg to about 3000mg per day, about 1000 mg to about 1400mg per day, or about 1200 mg per day.
  • the anti-insulin immunoglobulin preparation may be formulated for administration at a dose of about 5mg to about 25000mg per day, about 10 mg to about 20000 mg per day, about 25 mg to about 15000 mg per day, about 50 mg to about 10000 mg per day, about 50 mg to about 4000mg per day, about 500 mg to about 3000mg per day, about 1000 mg to about 1400mg per day or of about 1200 mg per day
  • the anti-insulin immunoglobulin preparation may be prepared by immunizing a mammal or avian with insulin conjugated to keyhole limpet hemocyanin (KLH).
  • KLH keyhole limpet hemocyanin
  • the present disclosure provides a method for reducing fasting glucose levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for increasing the early peak of insulin secretion in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing oral glucose tolerance in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for increasing insulin secretion in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing HBA1C levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing triglyceride levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing total cholesterol levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing LDL cholesterol levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing ALT levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing AST levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing ALP levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing GGT levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for increasing GLP-1 levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for increasing Adiponectin levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for increasing the Adiponectin/IL-6 ratio in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for increasing the CD25+ T regulatory cells in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing body weight in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing waist circumference or arm circumference in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in reducing fasting glucose levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in increasing the early peak of insulin secretion in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing oral glucose tolerance in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in increasing insulin secretion in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing HBA1C levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing triglyceride levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing total cholesterol levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing LDL cholesterol levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing ALT levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing AST levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing ALP levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing GGT levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in increasing GLP-1 levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in increasing Adiponectin levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in increasing the Adiponectin/IL-6 ratio in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in increasing the CD25+ T regulatory cells in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing body weight in a human patient in need thereof.
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing waist circumference or arm circumference in a human patient in need thereof.
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for reducing fasting glucose levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for increasing the early peak of insulin secretion in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing oral glucose tolerance in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for increasing insulin secretion in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing HBA1C levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing triglyceride levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing total cholesterol levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing LDL cholesterol levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing ALT levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing AST levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing ALP levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing GGT levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for increasing GLP-1 levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for increasing Adiponectin levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for increasing the Adiponectin/IL-6 ratio in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for increasing the CD25+ T regulatory cells in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing body weight in a human patient in need thereof.
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing waist circumference or arm circumference in a human patient in need thereof.
  • the present disclosure provides a method of treating a human suffering a T-cell mediated disease comprising administering to the human an effective amount of a composition comprising an anti-LPS immunoglobulin preparation
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in treating a human suffering a T-cell mediated disease
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for the treatment and/or prophylaxis of a human subject suffering a T-cell mediated disease
  • the T-cell mediated disease may be insulin resistance, impaired glucose tolerance, diabetes, metabolic syndrome, or a disease associated therewith, or non-alcoholic steatohepatitis (NASH).
  • NASH non-alcoholic steatohepatitis
  • the present disclosure provides a method of treatment of a human suffering a disease selected from insulin resistance or associated disorders comprising administering an effective amount of a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in treatment of a human suffering a disease selected from insulin resistance or associated disorders.
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament treatment of a human suffering a disease selected from insulin resistance or associated disorders.
  • the insulin resistance or an associated disorder may be diabetes, metabolic syndrome or non-alcoholic steatohepatitis (NASH).
  • diabetes metabolic syndrome or non-alcoholic steatohepatitis (NASH).
  • NASH non-alcoholic steatohepatitis
  • the anti-LPS immunoglobulin enriched colostrum preparation used in some embodiments of the methods described herein is referred to in the figures variously as Imm124-E, Travelan, Anti-LPS, T-IgG (produced by vaccination with multiple strains), and HIBC (produced by vaccination with a single strain).
  • a productive immune response results from the effective integration of positive and negative signals that have an impact on both innate and adaptive immune cells.
  • positive signals dominate, cell activation and pro-inflammatory responses ensue, resulting in the elimination of pathogenic microorganisms, viruses as well a transformed cell.
  • cell activation is blocked and active anti-inflammatory responses can occur.
  • Modulation of this binary system occurs through the action of cytokines, downstream signaling pathways and cell-cell contact. The perturbation of these thresholds can result in aberrant responses that are either insufficient to deal with pathogenic microorganisms or result in the loss of tolerance and the induction of autoimmune responses.
  • the present disclosure shows an immunomodulatory effect of a colostrum-derived immunoglobulin preparation enriched in anti-lipopolysaccharide (LPS) antibodies that may act in an active manner for the treatment of immune-related disorders.
  • LPS anti-lipopolysaccharide
  • Tregs are increasingly recognized as an important immunomodulatory component of the adaptive immune system. Immune dysregulation may lead to chronic inflammation as a trigger for chronic insulin insensitivity.
  • the present disclosure shows in a particular example, that oral administration of colostrum-derived anti-LPS antibodies promote Tregs in adipose tissue and in adipose tissue associated stromal vasculature. These alterations are associated with alleviation of the Metabolic Syndrome and liver injury in the ob/ob mice model. Therefore, the present disclosure provides as a novel therapeutic composition for the alleviation and treatment of the Metabolic Syndrome.
  • CD25+ LAP+ T cells, CD4+ CD25+ T cells, CD4+ CD25+ LAP- T cells, CD45+ LAP+ T and CD3+ LAP+ T cells are induced in the liver.
  • CD45+ LAP+ T cells, CD8+ LAP+ T cells, CD3+ LAP+ T, CD8+ CD25+ T cells are induced in the spleen.
  • CD4+ CD25+ T cells, CD3+ LAP+ T cells, CD4+ CD25+ LAP- T cells are induced in adipose tissue.
  • CD4+ CD25+ T cells and CD4+ CD25+ LAP+ T cells are induced in stromal vascular cells, CD3+ NK1.1+ cells in the liver, and CD25+ LAP- T cells are decreased in the liver.
  • adipokines Various constituents of the adipose tissue, such as mature adipocytes and stromal vascular cells, have distinct functions. They express and secrete different kinds of bioactive molecules collectively called adipokines. Altered adipokine secretion patterns characterize obesity and insulin resistance, which are major risk factors for type 2 diabetes mellitus. Regional and genotypic differences are present in stromal-vascular cells from obese and lean Zucker rats ( Turkenkopf, I. J. et al. Int. J. Obes. 12:515-24 (1988 )). Gene expression profiling using DNA microarrays showed differences between adipose tissue, adipocytes, and stromal vascular cells (Permana (2008) ibid.). The present disclosure further supports this notion, showing that the distribution of Tregs in these tissues is important in the metabolic syndrome and liver diseases.
  • the disclosure further shows that the promotion of Tregs in the adipose tissue and SV by administration of anti-LPS antibodies is associated with insulin resistance alleviation. This is demonstrated by glucose tolerance tests.
  • the inflammatory liver damage is alleviated by the present disclosure, as manifested by a decrease in liver enzymes.
  • the disclosure shows that oral administration of colostrum-enriched with anti-LPS antibodies can serve as a mean to promote Tregs in the adipose tissue and the adipose tissue associated stromal vasculature.
  • the disclosure also presents synergy between colostrum-derived components and anti-LPS antibodies by the effect on the distribution of Tregs.
  • Several proteins were identified in breast milk as involved in host defense ( Kahn, S. E. et al. Nature 444:840-6 (2006 )), including high concentrations mediators of the innate immune system ( Poggi, M. et al. Diabetologia (2009 )).
  • mediators include multiple defensin proteins, sphingolipids, osteopontin, exosomes, TLRs, cathelicidin, ⁇ eosinophil- derived neurotoxin, and high-mobility group box protein 1, and LL-37 (Poggi (2009) ibid.; Nagatomo, T.
  • BC bovine colostrum
  • BGS bovine colostrum
  • Induction of Treg cells may result in a long-lasting tolerance to ⁇ cell antigens, mediated by local immune modulation in the pancreatic draining lymph nodes (PLNs) ( Homann, D. et al. J. Immunol. 163:1833-8 (1999 ); Homann, D. et al. Immunity 11:463-72 (1999 )).
  • PPNs pancreatic draining lymph nodes
  • This intervention has shown great promise in animal models, but has had little efficacy in human trials.
  • In the Diabetes Prevention Trial only a sub-fraction of treated patients showed a beneficial effect with immunization with islet autoantigens ( Skyler, J.S. et al. Diabetes Care 28:1068-76 (2005 )).
  • the present inventors have shown dose dependent effects on the immune system.
  • the disclosure clearly demonstrates that anti-LPS antibodies together with colostrum adjuvants can promote Treg cell accumulation, and thereby serve as a means for alleviating inflammatory response, improving liver damage and improving Metabolic Syndrome complications.
  • Regulatory T lymphocytes in the adipose tissue and the SV can serve as a new therapeutic target in Metabolic Syndrome patients.
  • the immunoglobulins in the colostrum may promote regulatory T cells or any other cell related to the immune system in an antigen specific and non-specific way, by targeting bystander antigens, or by being directed against non-associated antigens.
  • the present disclosure provides a composition comprising an anti-LPS enriched immunoglobulin preparation for use in treatment and/or prophylaxis of a pathologic disorder.
  • the anti-LPS enriched immunoglobulin preparation may be derived from colostrum or from avian eggs.
  • Treatment refers to the reduction or elimination of the severity of a symptom of the disease, the frequency with which such a symptom is exhibited, or both.
  • “Prophylaxis” as used herein refers to completely or partially preventing or inhibiting a symptom of the disease or the frequency with which such a symptom is exhibited.
  • the present disclosure provides a composition for the treatment and prophylaxis of a pathologic disorder as described herein.
  • the composition of the disclosure comprises as active ingredient a mammalian anti-lipopolysaccharide (anti-LPS) enriched colostrum-derived immunoglobulin preparation or anti-LPS immunoglobulin preparation and optionally further colostrum, milk or milk product component/s, and any adjuvant/s.
  • anti-LPS mammalian anti-lipopolysaccharide
  • the immunoglobulin preparation or any fractions thereof recognizes and binds LPS and any fragments thereof.
  • the composition of the disclosure may comprise a combination of anti-LPS enriched colostrum-derived-immunoglobulin preparation with at least one immunoglobulin preparation comprising immunoglobulins recognizing at least one antigen specific for said disorder, thereby activating or inhibiting an immune response specifically directed towards said disorder.
  • the anti-LPS enriched colostrum-derived immunoglobulin preparations of the invention may be combined with any other immune modulatory drug, including but not limited to other colostrums derived antibodies, other antigen, other adjuvant, other cytokines or any type of molecule that can alter any component of the immune system.
  • the combination can be administered as one product, or in two or more separate products.
  • the combination may be administered together or separately from one another.
  • the methods described herein can be used for selecting a treatment for a subject.
  • the methods include detecting the presence in a sample from the subject of one or more of: an ALT level above 50 IU/dL (e.g., above 55, 60, 65, or 70, or alternatively above 40 or 45 IU/DL), an AST level above 50 IU/dL (e.g., above 55, 60, 65, or 70, or alternatively above 40 or 45 IU/DL), an AP of greater than 70 U/L (e.g., above 75, 80, 85, or 90, or alternatively above 60 or 65 IU/DL), a GGT of greater than 60 U/L (e.g., above 65, 70, 75, or 80, or alternatively above 50 or 55 IU/DL); a level of HbAlc above 7% (e.g., above 8, 9, 10, 11, 12, or 13%, or alternatively above 6.5 %), a level of fasting insulin secretion above 300 pg/ml (i
  • the subject has metabolic syndrome or non-alcoholic steatohepatitis (NASH); metabolic syndrome or diabetes; or elevated cholesterol levels.
  • the composition reduces AST and/or ALT levels; reduces serum insulin levels, increases serum insulin levels, improves fasting glucose levels, and/or reduces HbAlc levels; reduces serum cholesterol levels, reduces levels of serum LDL, and/or reduces levels serum triglycerides.
  • the colostrum-derived anti-LPS enriched immunoglobulin preparation or anti-LPS immunoglobulin preparation may comprise monomeric, dimeric or multimeric immunoglobulin selected from the group consisting of IgG, IgA and IgM and any fragments thereof.
  • the principal compositional difference between colostrum and mature milk is the very high content of colostral immunoglobulin, of which IgG class makes up 80-90%.
  • the colostrum-derived anti-LPS enriched immunoglobulin preparation or anti-LPS immunoglobulin preparation of the disclosure mainly comprises IgG, specifically, IgG1 and IgG2.
  • Immunoglobulin G is a multimeric immunoglobulin, built of two heavy chains and two light chains. Each complex has two antigen binding sites. This is the most abundant immunoglobulin and is approximately equally distributed in blood and in tissue liquids, constituting 75% of serum immunoglobulins in humans. In general, the number of IgG subclasses varied widely between different species, ranging from one subclass in rabbits to seven subclasses in horses, making it difficult to find orthologues. In humans, for example, IgG1 and IgG3 are the most pro-inflammatory IgG subclasses. In mice, however, IgG2a and IgG2b are the most pro-inflammatory IgG molecules showing a greater activity than mouse IgG1 and IgG3 in many in vivo model systems.
  • the anti-LPS enriched immunoglobulin preparation or anti-LPS immunoglobulin preparation may comprise a secretory antibody, specifically, sIgA.
  • IgA and IgM are secreted by a number of exocrine tissues.
  • IgA is the predominant secretory immunoglobulin present in colostrum, saliva, tears, bronchial secretions, nasal mucosa, prostatic fluid, vaginal secretions, and mucous secretions from the small intestine.
  • IgA output exceeds that of all other immunoglobulins, making it the major antibody produced by the body daily and is the major immunoglobulin found in human milk, whey and colostrum.
  • IgM secretion is less abundant but can increase to compensate for deficiencies in IgA secretion.
  • IgA J chain containing IgA is produced and secreted by plasma B immunocytes located in the lamina basement membrane of exocrine cells.
  • IgA has a typical immunoglobulin four-chain structure (M r 160,000) made up of two heavy chains (M r 55,000) and two light chains (M r 23,000).
  • M r 160,000 immunoglobulin four-chain structure
  • IgAl and IgA2 that have one and two heavy chains, respectively.
  • IgA can occur as monomers, dimers, trimers or multimers. In plasma, 10% of the total IgA is polymeric while the remaining 90% is monomeric.
  • the secreted IgA binds to a M r 100,000 poly-Ig receptor positioned in the basolateral surface of most mucosal cells.
  • the receptor-IgA complex is next translocated to the apical surface where IgA is secreted.
  • the binding of dimeric IgA to the poly-Ig receptor is completely dependent upon the presence of a J chain. Monomeric IgA will not bind to the receptor.
  • IgG The difference in function of lgG and IgA, follows the position where the molecules operate. IgA is found mainly on mucosal surfaces where there is little in the way of tissue fluid to carry immune cells and chemicals. Therefore, IgA (often as a dimer) would be preferably used for physical neutralisation of pathogens, and may be too effective at other immune functions. IgGs are present in the tissue fluid and blood where there is the full collection of leukocytes, complement system, macrophages etc.
  • phagocytes e.g., Killer T cells and macrophages
  • the anti-LPS enriched immunoglobulin preparations or anti-LPS immunoglobulin preparations of the disclosure may be obtained from any one of colostrum, colostrum serum, hyperimmunised milk or colostrum, colostrum whey (either cheese or casein), cheese or casein whey, directly from skim milk, whole milk, or a reconstituted form of such streams.
  • the anti-LPS enriched immunoglobulin preparation or anti-LPS immunoglobulin preparation comprised within the composition of the invention may be any fraction of colostrum.
  • colostrum where used herein includes colostral milk, processed colostral-milk such as colostral milk processed to partly or completely removes one or more of fat, cellular debris, lactose and casein.
  • the colostrum, or milk, containing the anti-LPS antibodies and optionally, the antigen-specific antibodies may be preferably collected by milking the animal colostrum or milk thus collected can either be used directly, may be further processed, for instance to purify anti-LPS and optionally, antigen-specific antibodies.
  • Methods for the (partial) purification of (LPS and optionally, antigen-specific) antibodies from colostrum or milk are present in the art.
  • any adjuvants may be added to the compositions of the invention.
  • Appropriate adjuvants therefore may be any antigen, antibody, glycosphingolipids, proteins, cytokines, adhesion molecules, and component that can activate or alter the function of antigen presenting cell or of any other cell related to the immune system in a direct and indirect manner.
  • the anti-LPS enriched immunoglobulin preparation or anti-LPS immunoglobulin preparation may be an affinity purified antibody or any fragment thereof.
  • antibody is meant to include both intact molecules as well as fragments thereof, such as, for example, Fab and F(ab') 2 , which are capable of binding antigen.
  • Fab and F(ab') 2 fragments lack the Fc fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding than an intact antibody. It will be appreciated that Fab and F(ab') 2 and other fragments of the antibodies useful in the present invention may be used for immuno-modulation, according to the methods disclosed herein for intact antibody molecules.
  • Such fragments are typically produced by proteolytic cleavage, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab') 2 fragments).
  • An antibody is said to be "capable of specifically recognizing" a certain antigen if it is capable of specifically reacting with an antigen which is in this particular example an antigen or a mixture of antigens specific for a certain immune-related disorder, to thereby bind the molecule to the antibody.
  • an "antigen” is a molecule or a portion of a molecule capable of being bound by an antibody, which is additionally capable of inducing an animal to produce antibody capable of binding to an epitope of that antigen.
  • An antigen may have one or more than one epitope.
  • epitope' t is meant to refer to that portion of any molecule capable of being bound by an antibody that can also be recognized by that antibody.
  • Epitopes or "antigenic determinants” usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains, and have specific three-dimensional structural characteristics as well as specific charge characteristics.
  • the anti-LPS enriched immunoglobulin preparation or anti-LPS immunoglobulin preparation used as an active ingredient for the composition of the invention may be obtained from a mammal immunized with LPS or any fragments thereof.
  • said mammal in addition to LPS, may be further immunized with at least one antigen or a mixture of at least two antigens specific for said disorder, as well as with a mixture of at least two different antibodies directed against at least two different antigens associated with the disease.
  • the LPS or any antigen used for immunizing said mammal may be provided as any one of an isolated and purified peptide, a purified recombinant protein, a fusion protein, cell lysate, membranal preparation, nuclear preparation, or cytosolic preparation of any one of tissue culture cells, primary cells or tissue samples obtained from a subject suffering from said disorder.
  • the composition of the invention may optionally further comprise colostrum component/s such as for example, alarmins, defenensins, colostrinin, and any other colostrum or milk derived carbohydrates, glycolipids or any other molecules or components that may further enhance or inhibit modulation of an immune response, or any preparations, mixtures or combinations thereof.
  • the composition of the invention may comprise any additional adjuvant.
  • Appropriate adjuvants therefore may be any antigen, antibody, glycosphingolipids, proteins, cytokines, adhesion molecules, and component that can activate or alter the function of antigen presenting cell or of any other cell related to the immune system in a direct and indirect manner.
  • the composition comprises a constituent of a bird's egg, wherein the bird's egg comprises IgY specific for LPS or a fragment thereof.
  • Crude egg yolk may be used as an antibody source However, avian antibodies are usually purified or concentrated from the yolk prior to use.
  • the constituent of the bird's egg may be concentrated or purified as necessary, as is understood by those skilled in the art.
  • the composition comprises the yolk of the egg, or any IgY antibody-containing fraction thereof.
  • the yolk is preferable to the white of the egg, as the yolk typically contains much higher concentrations of IgY than does the white. However, the white may contain concentrations of IgY sufficient for some applications.
  • the IgY is concentrated, isolated, or purified from the constituent of the bird egg This can be accomplished by a variety of methods
  • the antibodies may be purified by the water dilution method.
  • the precipitate may then be removed by any conventional method, including centrifugation.
  • the supernatant can then be stored frozen, for example at -20° C.
  • IgY can then be isolated by precipitation with ammonium sulfate and subsequent dialysis.
  • the titer of IgY antibodies can be determined by immunoassay, for example ELISA.
  • the water dilution method is more completely described in the well-known literature, for example by Akita and Nakai (1993), which teach this method.
  • the antibody composition are substantially isolated.
  • a significant fraction of a non-antibody yolk component has been removed.
  • the non- antibody yolk component may be for example the lipid component of the yolk, the carbohydrate component of the yolk, the yolk granules, the hydrophobic component of the yolk, the steroid component of the yolk, and the non-immunoglobulin protein component of the yolk.
  • the fraction of the component removed is at least 50%. In some embodiments the removed fraction is at least 60%, 75%, 80%, 90%, 95%, 99%, or 99 9%. Greater removed fractions have the advantage of producing a more pure antibody composition. Smaller removed fractions have the advantage of requiring less processing.
  • the antibody composition are substantially concentrated. In such embodiments the concentration of IgY will be greater in the composition than in the egg yolk.
  • Substantially concentrated antibody compositions comprise IgY that is at least twice as concentrated as in the yolk.
  • Some embodiments of the substantially concentrated antibody composition are concentrated by at least a factor of 3, 4, 5, 6, 7, 8, 9, 10, 100, 1000, or 10,000. More concentrated antibody compositions have the advantage of providing the same mass of antibodies in lower volume. Less concentrated antibody compositions have the advantage of requiring less processing.
  • the antibody compositions of the present disclosure may be processed so as to largely remove all isotypes except IgG and IgY.
  • the immunoglobulin may be derived from numerous donors. Any number of donors may be used
  • the antibodies are derived from one donor.
  • the antibodies are derived from 1-10 donors.
  • the antibodies are derived from 10-100 donors.
  • the antibodies are derived from 100-1000 donors.
  • the antibodies are derived from over 1000 donors.
  • the composition is made by the method comprising obtaining an egg laid by a fowl previously immunized against influenza and separating the antibody fraction from a yolk of the egg.
  • the fowl has been actively immunized, for example by vaccination.
  • the fowl is preferably a domesticated fowl
  • the domesticated fowl may be chicken, duck, swan, goose, turkey, peacock, guinea hen, ostrich, pigeon, quail, pheasant, dove, or other domesticated fowl
  • the domesticated fowl is preferably a chicken
  • the domesticated fowl is more preferably a domesticated chicken raised primarily for egg or meat production.
  • the fowl may be immunized against any strain of influenza, any subtype of influenza, any type of influenza, or combinations thereof.
  • the antibody composition is made by a method comprising actively immunizing a hen with antigen, collecting eggs from the hen after an immunization period, and separating the antibody fraction from a yolk of the egg.
  • collecting eggs from the hen can occur continuously after the immunization period.
  • the immunization of the bird may occur by any means known in the art.
  • a vaccine may be administered to the bird that is known to effectively elicit an immune response in birds, or that is known to effectively elicit an immune response in mammals.
  • Many such influenza vaccines are commercially available, and can be routinely developed by those of ordinary skill m the art without undue experimentation further methods of producing IgY with a specific target are known to those skilled in the art.
  • the present disclosure provides a composition comprising an anti-LPS enriched immunoglobulin preparation or anti-LPS immunoglobulin preparation for use in treatment and/or prophylaxis of a pathologic disorder wherein the anti-LPS enriched immunoglobulin preparation is derived from avian eggs and further comprising non-hyperimmune colostrum.
  • the pathologic disorder is acute or chronic liver disease, cirrhosis or any disease or complication associated therewith.
  • the acute or chronic liver disease, cirrhosis and any disease or complication associated therewith is selected from the group consisting of hepatic encephalopathy, spontaneous bacterial peritonitis (SBP), ascites, bleeding varices, cirrhosis associated hyperdynamic circulation, hepatorenal syndrome, hepatopulmonary syndrome, portopulmonary hypertension, variceal bleeding, adrenal insufficiency and altered level of consciousness.
  • the pathologic disorder is liver damage.
  • the pathologic disorder is an immune-related disorder selected from the group consisting of autoimmune disease, non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith, infectious disease, and proliferative disorder.
  • the pathologic disorder may be selected from the group consisting of secondary peritonitis and infection after surgery, hepatic cardiomyopathy and hypotension, hepatoadrenal syndrome, hepatocellular carcinoma, Alzheimer's disease, any type of memory loss, any type of dementia, attention deficit disorders (ADHA), any type of learning disability, effect of alcohol or drugs on the brain, any type of immune mediated disease including asthma, and peritonitis.
  • the immuno-modulating composition of the disclosure is capable of reducing, eliminating or inhibiting mucosal microbial translocation, thereby modulating immune activation.
  • chronic activation of the immune system is a hallmark of progressive viral infection and predicts disease outcome.
  • circulating microbial products likely derived from the gastrointestinal tract, in a process also known as "mucosal microbial translocation”
  • the compositions of the disclosure may modulate immune function, or alternatively, reduce or change the number of bacteria or of bacteria related products not related to alteration of the immune system.
  • the disclosure provides a composition
  • a composition comprising as an active ingredient a mammalian anti-lipopolysaccharide (LPS) enriched colostrum-derived immunoglobulin preparation.
  • LPS mammalian anti-lipopolysaccharide
  • Such composition wherein said composition is particularly applicable for the treatment, prevention and prophylaxis of acute or chronic liver disease, cirrhosis and any disease or complication associated therewith, optionally said composition further comprises an additional therapeutic agent or any carrier and adjuvant.
  • liver disease may be for example, at least one of hepatic encephalopathy, spontaneous bacterial peritonitis (SBP), ascites, variceal bleeding, cirrhosis associated hyperdynamic circulation, hepatorenal syndrome, hepatopulmonary syndrome, portopulmonary hypertension, variceal bleeding, adrenal insufficiency and altered level of consciousness.
  • SBP spontaneous bacterial peritonitis
  • composition of the disclosure may be used for the treatment of pathologic disorders such as any type of viral disease including HCV, HBV, CMV, and EBV.
  • colostrum-derived preparations may be therefore combined with any drug used for liver disease, as an additional therapeutic agent.
  • Cirrhosis refers to the final common histological outcome of a wide verity of chronic liver diseases, characterized by the replacement of liver tissue by fibrous scar tissue and regeneration of nodules, leading to progressive loss of liver function. Cirrhosis is usually caused by Hepatitis B and C viruses, alcoholism and fatty liver disease.
  • cites describes the condition of pathologic fluid accumulation within the abdominal cavity, most commonly due to cirrhosis and sever liver disease.
  • colostrum-derived preparations may be therefore combined with any immunomodulatory therapeutic agent/s or any combination or mixture thereof, creating a combined immunomodulatory composition for the treatment and/or prevention of immune-related disorders, a non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith, infectious disease, malignant or infectious disorders.
  • colostrum-derived composition of the invention may further comprises any added adjuvant.
  • the disclosure further provides the use of the anti LPS compositions of the disclosure, optionally, combined with colostrum preparations enriched for antibodies directed against antigens associated with a disease, for example, anti-insulin antibodies, in the treatment of any acute or chronic liver disease, diabetes, and any complication of diabetes, fatty liver, non-alcoholic steatohepatitis, and obesity.
  • the composition further comprises an immunoglobulin preparation comprising immunoglobulins that recognize and bind at least one antigen specific for said pathologic disorder.
  • the further immunoglobulin preparation may be derived from colostrum or from avian eggs.
  • the disclosure provides combined compositions comprising a combination of anti-LPS enriched immunoglobulin preparation with at least one colostrum-derived immunoglobulin preparation comprising immunoglobulins that recognize and bind at least one antigen specific for said pathologic disorder and thereby modulate immune-regulatory cells, specifically, regulatory T cells.
  • modulation may results for example, in modulation of the Th1/Th2, Trl/Th3 cell balance toward an anti-inflammatory Th2, Trl/Th3 immune response thereby inhibiting an immune response specifically directed toward said disorder.
  • Immunoglobulins that recognize and bind at least one antigen specific for said pathologic disorder and thereby modulate immune-regulatory cells, specifically, regulatory T cells include the following:
  • the anti-LPS enriched immunoglobulin preparation of the invention may further comprise immunoglobulins directed to antigens that are not specific to the treated disorder.
  • antigens may be any target immune-related components having a modulatory effect on the immune-response.
  • recognition of such disease non-specific antigens by the immunoglobulin preparation of the invention may results in alteration of the immune-response.
  • modulation may results for example, in modulation of the Th1/Th2, Trl/Th3 cell balance toward an anti-inflammatory Th2, Trl/Th3 immune response thereby inhibiting an immune response specifically directed toward said disorder.
  • the combined composition of the invention may optionally further comprises an additional therapeutic agent or any carrier and adjuvant.
  • the combined colostrums-derived immunoglobulin preparation of the invention as well as the immuno-modulatory composition derived therefrom may act in an indirect manner by activation or promotion of specific subsets of regulatory cells, or antigen presenting cells, or by any type of cell-cell contact.
  • Such anti-LPS enriched combined composition may be directed towards different components of the immune-system. For example, activation of specific regulatory T cells, B cells or antigen presenting cells, or any other cells that associated with an effect on the immune system, or induces the secretion of cytokines or chemokines or affects the immune system in any other way.
  • Alteration or promotion of immune cells may further involve induction of any type of regulatory cells, preferably, regulatory T cells, for example, Th3 cells, Trl, T17 cells or any other type of regulatory, effector or suppressor cells.
  • regulatory T cells for example, Th3 cells, Trl, T17 cells or any other type of regulatory, effector or suppressor cells.
  • Th17 cells are a recently-identified subset of CD4 T helper cells. They are found at the interfaces between the external environment and the internal environment, e.g., skin and lining of the GI tract.
  • the colostrum-derived anti-LPS enriched immunoglobulin preparations of the invention may promote regulatory T cells or any other cell related to the immune system in an antigen specific and non specific manner, by targeting bystander antigens, or by being directed towards non associated antigens.
  • the invention provides a combination of an anti-LPS enriched immunoglobulin preparation of the invention with at least one additional immunoglobulin preparation comprising immunoglobulins directed against at least one antigen associated with said disorder, creating a combined composition for treating immune-related disorders.
  • Such composition therefore may be antigen or disease specific or alternatively, may augment or induce specific cells or parts of the immune system in a non-antigen specific way, including an immune bystander effect.
  • the composition modulates regulatory T cells leading to modulation of the Th1/Th2, Trl/Th3 cell balance toward an anti-inflammatory Th2, Trl/Th3 immune response or a pro-inflammatory Th1 immune response thereby inhibiting or activating an immune response specifically directed toward said disorder.
  • the composition modulates the Th1/Th2, Trl/Th3 cell balance toward an anti-inflammatory Th2, Trl/Th3 immune response thereby inhibiting an immune response specifically directed toward said disorder, and wherein said composition is for the treatment of any one of an autoimmune disease, non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith selected from diabetes type 2, insulin resistance, obesity and overweight.
  • the composition is for the treatment and/or prophylaxis of metabolic syndrome or non-alcoholic steatohepatitis or both.
  • the composition is for the treatment, and/or prophylaxis of diabetes, the treatment of impaired glucose tolerance, such as decreasing glucose tolerance. decreasing serum insulin levels, decreasing hepatic triglyceride levels, or decreasing cholesterol levels.
  • the composition modulates the Th1/Th2, Trl/Th3 cell balance toward a pro-inflammatory Th1/Th2 immune response thereby enhancing an immune response specifically directed toward said disorder, and wherein said composition is for the treatment of infectious diseases, and proliferative disorders.
  • composition may further comprise a therapeutic agent, carrier or adjuvant and/or non-hyperimmune colostrum.
  • anti-LPS enriched immunoglobulin preparation of the invention may be used either for an active or a passive treatment.
  • said immune-related disorder is any one of autoimmune disease, infectious disease, and proliferative disorder.
  • composition of the disclosure may be applicable for treating acute complications, or prevention the development or the recurrence of these complications.
  • the combined composition of the disclosure leads to modulation of the Th1/Th2, Trl/Th3 cell balance toward an anti-inflammatory Th2, Trl/Th3 immune response thereby inhibiting an immune response specifically directed toward said disorder.
  • Such regulation may involve regulatory T cells, antigen presenting cells, any type of T cell or B cell, the function of any cell associated directly or indirectly with the immune system, or any type of cytokine or chemokine, or adjuvant.
  • such composition may be applicable in the treatment of an autoimmune disease.
  • autoimmune disorders include, but are not limited to, Alopecia Areata, Lupus, Anlcylosing Spondylitis, Meniere's Disease, Antiphospholipid Syndrome, Mixed Connective Tissue Disease, Autoimmune Addison's Disease, Multiple Sclerosis, Autoimmune Hemolytic Anemia, Myasthenia Gravis, Autoimmune Hepatitis, Pemphigus Vulgaris, Behcet's Disease, Pernicious Anemia, Bullous Pemphigoid, Polyarthritis Nodosa, Cardiomyopathy, Polychondritis, Celiac Sprue-Dermatitis, Polyglandular Syndromes, Chronic Fatigue Syndrome (CFIDS), Polymyalgia Rheumatica, Chronic Inflammatory Demyelinating, Polymyositis and Dermatomyositis, Chronic Inflammatory Polyneuropathy, Primary Agammaglobulinemia, Churg-Strauss Syndrome, Primary Biliary Cirrhosis, Cicatricial Pe
  • compositions described herein can be administered to a subject to treat or prevent disorders associated with an abnormal or unwanted immune response associated with cell, tissue or organ transplantation, e.g., renal, hepatic, and cardiac transplantation, e.g., graft versus host disease (GVHD), or to prevent allograft rejection.
  • disorders associated with an abnormal or unwanted immune response associated with cell, tissue or organ transplantation e.g., renal, hepatic, and cardiac transplantation, e.g., graft versus host disease (GVHD), or to prevent allograft rejection.
  • GVHD graft versus host disease
  • compositions of the disclosure may be used for treating any one of non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith for example, diabetes type 2, insulin resistance, obesity and overweight.
  • the combined composition of the disclosure may lead to modulation of the Th1/Th2, Trl/Th3 cell balance toward a pro-inflammatory Th1/Th2 immune response thereby enhancing an immune response specifically directed toward said disorder.
  • Such regulation may involve regulatory T cells, antigen presenting cells, any type of T cell or B cell, the function of any cell associated directly or indirectly with the immune system, or any type of cytokine or chemokine, or adjuvant.
  • such composition may be applicable in the treatment of infectious diseases, and proliferative disorders.
  • a malignant proliferative disorder may be a solid or non-solid tumor, for example, carcinoma, sarcoma, melanoma, leukemia, myeloma or lymphoma.
  • the composition of the disclosure is intended for preventing and/or treating carcinoma such as hepaotcellular carcinoma, prostate cancer, breast carcinoma, colon carcinoma.
  • the composition of the disclosure may be used for preventing and/or treating leukemia, more specifically, acute or chronic leukemia.
  • cancer As used herein to describe the present disclosure, “cancer”, “tumor” and “malignancy” all relate equivalently to a hyperplasia of a tissue or organ. If the tissue is a part of the lymphatic or immune systems, malignant cells may include non-solid tumors of circulating cells. Malignancies of other tissues or organs may produce solid tumors. In general, the methods and compositions of the present disclosure may be used in the treatment of non-solid and solid tumors.
  • Malignancy as contemplated in the present disclosure may be selected from the group consisting of carcinomas, melanomas, lymphomas and sarcomas.
  • Malignancies that may find utility in the present disclosure can comprise but are not limited to hematological malignancies (including leukemia, lymphoma and myeloproliferative disorders), hypoplastic and aplastic anemia (both virally induced and idiopathic), myelodysplastic syndromes, all types of paraneoplastic syndromes (both immune mediated and idiopathic) and solid tumors (including lung, liver, breast, colon, prostate GI tract, pancreas and Karposi). More particularly, the malignant disorder may be hepaotcellular carcinoma, colon cancer, melanoma, myeloma and acute or chronic leukemia.
  • the immuno-modulating composition of the disclosure may be specifically applicable for treating infectious diseases, for example, conditions caused by viral pathogens such as HCV, HBV, CMV, and EBV.
  • infectious diseases for example, conditions caused by viral pathogens such as HCV, HBV, CMV, and EBV.
  • the combined immunomodulatory composition of the disclosure may lead to a Th2, Trl/Th3 anti-inflammatory response. More specifically, such anti-inflammatory response may be accompanied by a decrease or reduction in the amount or expression of pro-inflammatory cytokines such as IL-2, IL-17, IL-23, IFN- ⁇ , IL-6.
  • Such decrease or reduction according to the disclosure may be a reduction of about 5% to 99%, specifically, a reduction of about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 99% as compared to untreated control.
  • the composition of the disclosure may elevate and increase the amount or expression of anti-inflammatory cytokines such as TGF- ⁇ , IL-10, IL-4, IL-5, IL-9 and IL-13. More specifically, the increase, induction or elevation of the anti-inflammatory cytokines may be an increase of about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 99% as compared to untreated control.
  • anti-inflammatory cytokines such as TGF- ⁇ , IL-10, IL-4, IL-5, IL-9 and IL-13. More specifically, the increase, induction or elevation of the anti-inflammatory cytokines may be an increase of about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 99% as compared to untreated control.
  • the anti-inflammatory effect of the combined immuno-modulatory composition of the disclosure may be achieved by activation or promotion of specific subsets of regulatory cells, antigen presenting cells or any type of cell-cell contact, or via direct or indirect activation of cytokines and/or chemokines. It should be further noted that any type of regulatory or effector cell, specifically regulatory T cells, including Th3 and Trl. cells may be involved.
  • the colostrum-derived anti-LPS enriched immunoglobulin preparations of the disclosure may promote regulatory T cells or any other cell related to the immune system in an antigen specific and non specific way, by targeting bystander antigens, or by being directed against non associated antigens.
  • an immune-related cell activated or promoted by the composition of the disclosure may be an APC (such as DC), Treg cell or any other cell associated directly on indirectly with the immune system including but not limited to platelets, macrophages, any type of B cell, T cell (including double negative cells), and any type of non-professional antigen presenting cell, adipocytes, endothelial cell, any type of cell that is part of an organ, specifically, an organ connected to the treated immune-related disorder and any type of cell having regulatory enhancing or suppressing properties. More particularly, the compositions of the disclosure demonstrate anti-inflammatory effect on immune-related cells such as specific T regulatory cells for example, adipocytes and Antigen Presenting Cells (APC), such as DC.
  • APC Antigen Presenting Cells
  • the composition of the disclosure may be used for inducing at least one of T regulatory (Treg) cells, or any cell having regulatory properties, either suppressive or inductive, adipocyte and Antigen Presenting Cells (APC) in a subject suffering from hepatic disorder.
  • T regulatory T regulatory
  • APC Antigen Presenting Cells
  • compositions or the optional combined compositions of the disclosure are intended for preventing and/or treating a pathologic disorder, specifically, hepatic disorders, or an immune-related disorder.
  • disorder refers to a condition in which there is a disturbance of normal functioning.
  • a “disease” is any abnormal condition of the body or mind that causes discomfort, dysfunction, or distress to the person affected or those in contact with the person. Sometimes the term is used broadly to include injuries, disabilities, syndromes, symptoms, deviant behaviors, and atypical variations of structure and function, while in other contexts these may be considered distinguishable categories. It should be noted that the terms “disease”, “disorder”, “condition” and “illness”, are equally used herein.
  • an "immune-related disorder or disease” or “hepatic disorder” may be any disorder associated with, caused by, linked to, a non normal immune response. Such disorders may usually occur together with a disturbed immune response, or believed to have an impact on or by a non normal immune response.
  • composition may be formulated for administration orally, by inhalation as an aerosol, or by parenteral, intravaginal, intranasal, mucosal, sublingual, topical, or rectal administration, or any combination thereof.
  • the immunoglobulin preparation or any fractions thereof recognizes and binds LPS or any fragments thereof.
  • the composition inhibits microbial translocation. In another embodiment, the composition inhibits microbial translocation and thereby modulates immune activation.
  • the present provides a composition comprising a mammalian anti-LPS enriched colostrum-derived immunoglobulin preparation for modulating immune tolerance in a subject, or in another aspect, for modulating oral tolerance in a subject
  • any of the compositions of the disclosure may be administered orally or by inhalation as an aerosol or by intravenous, intramuscular, subcutaneous, intraperitoneal, parenteral, transdermal, intravaginal, intranasal, mucosal, sublingual, topical, rectal or subcutaneous administration, or any combination thereof.
  • Orally administrated antibodies would be expected to be degraded in the gastrointestinal tract, given the low gastric pH and the presence of gastric and intestinal proteases.
  • bovine colostral IgG (BCIg) has been cited as particularly resistant to GI destruction, relative to other immunoglobulins.
  • BCIg bovine colostral IgG
  • Early studies of BCIg cited remarkable "resistance to proteolytic digestion in the intestine of a heterologous host".
  • bovine IgG1 is somewhat more resistant to proteolysis by trypsin, chymotrypsin and pepsin than other Igs.
  • the composition of the disclosure may be suitable for mucosal administration, for example, pulmonary, buccal, nasal, intranasal, sublingual, rectal, vaginal administration and any combination thereof.
  • any other route of administration may be applicable, for example, intravenous, intravenous, intramuscular, subcutaneous, intraperitoneal, parenteral, intravaginal, intranasal, mucosal, sublingual, topical, rectal or subcutaneous administration, or any combination thereof.
  • the anti-LPS enriched immunoglobulin preparation used by the compositions of the invention may be prepared in preparations such as food additives, aqueous solutions, oily preparations, emulsions, gels, etc., and these preparations may be administered orally, topically, rectally, nasally, bucally, or vaginally.
  • the preparations may be administered in dosage formulations containing conventional non-toxic acceptable carriers and may also include one or more acceptable additives, including acceptable salts, polymers, solvents, buffers, excipients, bulking agents, diluents, excipients, suspending agents, lubricating agents, adjuvants, vehicles, deliver systems, emulsifiers, dis-integrants, absorbents, preservatives, surfactants, colorants, flavorants or sweeteners.
  • An optional dosage form of the present invention may be a powder for incorporation into beverages, pills, syrup, capsules, tablets, granules, beads, chewable lozenges or food additives, using techniques known in the art.
  • immuno-modulating composition of the invention may be administered in a form selected from the group consisting of orally-active powders, pills, capsules, teas, extracts, dried extracts, subliguals, sprays, dispersions, solutions, suspensions, emulsions, foams, syrups ,lotions, ointments, gels, pastes, dermal pathces, injectables, vaginal creams and suppositories.
  • Therapeutic formulations may be administered in any conventional dosage formulation.
  • Formulations typically comprise at least one active ingredient, as defined above, together with one or more acceptable carriers thereof.
  • Each carrier should be both pharmaceutically and physiologically acceptable in the sense of being compatible with the other ingredients and not injurious to the patient.
  • Formulations include those suitable for oral, rectal, nasal, or parenteral (including subcutaneous, intramuscular, intravenous and intradermal or by inhalation) administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The nature, availability and sources, and the administration of all such compounds including the effective amounts necessary to produce desirable effects in a subject are well known in the art and need not be further described herein.
  • compositions are well known in the art and has been described in many articles and textbooks, see e.g., Remington's Pharmaceutical Sciences, Gennaro A. R. ed., Mack Publishing Co., Easton, PA, 1990, and especially pp. 1521-1712 therein.
  • composition of the invention can be administered and dosed in accordance with good medical practice.
  • composition of the invention may comprise the active substance in free form and be administered directly to the subject to be treated,
  • Formulations typically comprise at least one active ingredient, as defined above, together with one or more acceptable carriers thereof.
  • Each carrier should be both pharmaceutically and physiologically acceptable in the sense of being compatible with the other ingredients and not injurious to the patient.
  • Formulations include those suitable for oral, nasal, or par enteral (including subcutaneous (s.c.), intramuscular (i.m.), intraperitoneal (i.p.), intravenous (i.v.) and intradermal or by inhalation to the lung mucosa) administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The nature, availability and sources, and the administration of all such compounds including the effective amounts necessary to produce desirable effects in a subject are well known in the art and need not be further described herein.
  • compositions of the invention generally comprise a buffering agent, an agent that adjusts the osmolarity thereof, and optionally, one or more pharmaceutically acceptable carriers, excipients and/or additives as known in the art.
  • Supplementary active ingredients can also be incorporated into the compositions.
  • the carrier can be solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents and the like.
  • the use of such media and agents for pharmaceutical active substances is well known in the art. Except as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic composition is contemplated.
  • the active drug components can be combined with a non-toxic pharmaceutically acceptable inert carrier such as lactose, starch, sucrose, glucose, modified sugars, modified starches, methylcellulose and its derivatives, dicalcium phosphate, calcium sulfate, mannitol, sorbitol, and other reducing and non-reducing sugars, magnesium stearate, stearic acid, sodium stearyl fumarate, glyceryl behenate, calcium stearate and the like.
  • a non-toxic pharmaceutically acceptable inert carrier such as lactose, starch, sucrose, glucose, modified sugars, modified starches, methylcellulose and its derivatives, dicalcium phosphate, calcium sulfate, mannitol, sorbitol, and other reducing and non-reducing sugars, magnesium stearate, stearic acid, sodium stearyl fumarate, glyceryl behenate, calcium stearate and the like.
  • the active drug components can be combined with non-toxic pharmaceutically acceptable inert carriers such as ethanol, glycerol, water and the like.
  • suitable binders, lubricants, disintegrating agents and coloring and flavoring agents can also be incorporated into the mixture.
  • Stabilizing agents such as antioxidants, propyl gallate, sodium ascorbate, citric acid, calcium metabisulphite, hydroquinone, and 7-hydroxycoumarin can also be added to stabilize the dosage forms.
  • Other suitable compounds can include gelatin, sweeteners, natural and synthetic gums such as acacia, tragacanth, or alginates, carboxymethylcellulose, polyethylene, glycol, waxes and the like.
  • the present disclosure provides the use of a mammalian anti-LPS enriched colostrum-derived immunoglobulin preparation and optionally of a colostrum-derived immunoglobulin preparation recognizing at least one antigen specific for a pathologic disorder in the manufacture of an immuno-modulating composition for the treatment and prophylaxis of a pathologic disorder.
  • the anti-LPS enriched immunoglobulin preparation or any fractions thereof recognizes and binds LPS and any fragments thereof.
  • the composition prepared by the use of the invention may comprise a combination of the anti-LPS enriched immunoglobulin preparation of the invention and at least one immunoglobulin preparation comprising immunoglobulins recognizing at least one antigen specific for said disorder.
  • Such recognition leads to alteration of regulatory T cells, and as a result, causes modulation of the Th1/Th2, Trl/Th3 cell balance either toward an anti-inflammatory Th2, Trl/Th3 immune response or toward a pro-inflammatory Th1 immune response.
  • Trl/Th3 cell balance either toward an anti-inflammatory Th2, Trl/Th3 immune response or toward a pro-inflammatory Th1 immune response.
  • any type of regulatory or effector cells specifically regulatory T cells, including Th3 and Trl (T H 3, T cells are preferentially induced at mucosal surfaces and secrete transforming growth factor (TGF)- ⁇ ) cells may be involved.
  • TGF transforming growth factor
  • the colostrum-derived anti-LPS enriched immunoglobulin preparations of the invention may promote regulatory T cells or any other cell related to the immune system in an antigen specific and non specific way, by targeting bystander antigens, or by being directed against non associated antigens.
  • the anti-LPS enriched colostrum-derived immunoglobulin preparation used for the invention comprises monomeric, dimeric or multimeric immunoglobulin selected from the group consisting of IgG, IgA and IgM and any fragments, mixtures or combinations thereof.
  • the use according to the invention of colostrum-derived, milk or milk products-derived anti-LPS enriched immunoglobulin preparation is for manufacturing a composition or combined composition that optionally may further comprises colostrum, milk or milk products component/s and any adjuvant/s, preferably, alarmins, defenensins, colostrinin and any preparation, mixture or combination thereof.
  • the composition of the invention may comprise any additional adjuvant.
  • Appropriate adjuvants therefore may be any antigen, antibody, glycosphingolipids, proteins, cytokines, adhesion molecules, and component that can activate or alter the function of antigen presenting cell or of any other cell related to the immune system in a direct and indirect manner.
  • the present invention further provides the use of colostrum or any colostrum-derived preparations in the combined compositions of the invention for enhancing an immunomodulatory effect of an immunomodulatory therapeutic agent.
  • alarmin denotes an array of structurally diverse multifunctional host proteins that are rapidly released during infection or tissue damage, and that have mobilizing and activating effects on receptor-expressing cells engaged in host defence and tissue repair.
  • Innate-immune mediators that have alarmin function include defensins, eosinophil-derived neurotoxin, cathelicidins and HMGB 1.
  • Defensins are small (15-20 residue) cysteine-rich cationic proteins found in both vertebrates and invertebrates. They are active against bacteria, fungi and enveloped viruses. They consist of 15-20 amino acids including six to eight conserved cysteine residues. Cells of the immune system contain these peptides to assist in killing phagocytized bacteria, for example in neutrophil granulocytes and almost all epithelial cells. Most defensins function by penetrating the microbial's cell membrane by way of electrical attraction, and once embedded, forming a pore in the membrane which allows efflux.
  • Colostrinin refers to a polypeptide which, in its natural form, is obtained from mammalian colostrum. Colostrinin is sometimes known as “colostrinine", and has a molecular weight in the range 16,000 to 26,000 Daltons. Colostrinin may form a dimer or trimer of sub-units (each having a molecular weight in the range 5,000 to 10,000 Daltons, preferably 6,000 Daltons), and contains mostly praline (the amount of proline is greater than the amount of any other single amino acid).
  • Colostrinin is characterized in that it stimulates the production of cytokines, especially gamma interferon (IFN- ⁇ ), tumor necrosis factor TNF- ⁇ ), interleukins (e.g. IL-6 and IL-10) and various growth factors.
  • cytokines especially gamma interferon (IFN- ⁇ ), tumor necrosis factor TNF- ⁇ ), interleukins (e.g. IL-6 and IL-10) and various growth factors.
  • the anti-LPS enriched immunoglobulin preparation and any other optional immunoglobulin preparations used by the invention may be obtained from a mammal, immunized with LPS or any fragments thereof and optionally, in addition, with at least one antigen or a mixture of at least two antigens specific for the disorder to be treated.
  • Means and methods of the invention are suited to obtain high and prolonged antigen-specific antibody production in the colostrum, milk or milk products of any lactating mammal.
  • said animal is a farm-animal. Farm animals are animals that are used on a commercial basis by man, be it for the production of milk, meat or even antibodies.
  • Farm-animals already used for the commercial scale production of milk are preferred for the present invention since for these animals special lines and/or breeds exist that are optimized for milk production.
  • said farm-animal is a cow or a goat. More preferably said farm-animal is a cow.
  • the composition reduces or inhibits mucosal microbial translocation. In one embodiment of said use of the invention, the composition reduces or inhibits mucosal microbial translocation and thereby modulates immune activation.
  • the disclosure relates to the use of a mammalian anti-LPS enriched colostrum-derived immunoglobulin preparation for manufacturing a composition for the treatment, prevention and prophylaxis of acute or chronic liver disease, cirrhosis and any disease or complication associated therewith, optionally said composition further comprises an additional therapeutic agent or any carrier and adjuvant.
  • this particular composition reduces or inhibits mucosal microbial translocation and thereby alters the direct effect of bacteria or any other infectious agent on the pathogenesis of complications of acute or chronic liver diseases-associated complications whether due to portal hypertension or any other cause.
  • liver disease cirrhosis and any disease or complication associated therewith is at least one of hepatic encephalopathy, spontaneous bacterial peritonitis (SBP), ascites, cirrhosis associated hyperdynamic circulation, hepatorenal syndrome, hepatopulmonary syndrome, portopulmonary hypertension, variceal bleeding, adrenal insufficiency and altered level of consciousness.
  • SBP spontaneous bacterial peritonitis
  • the disclosure provides the use of a combination of anti-LPS enriched immunoglobulin preparation with at least one colostrum-derived immunoglobulin preparation comprising immunoglobulins that recognize and bind at least one antigen specific for said pathologic disorder.
  • the use of such combination is for preparing an immuno-modulatory composition that modulates regulatory T cells leading to modulation of the Th1/Th2, Tr1/Th3 cell balance toward an anti-inflammatory Th2, Tr1/Th3 immune response or a pro-inflammatory Th1 immune response thereby inhibiting or activating an immune response specifically directed toward said disorder.
  • such combined composition further comprises an additional therapeutic agent or any carrier and adjuvant.
  • Such composition modulates regulatory T cells leading to modulation of the Th1/Th2, Tr1/Th3 cell balance toward an anti-inflammatory Th2, Tr1/Th3 immune response or a pro-inflammatory Th1 immune response, thereby inhibiting or activating an immune response specifically directed toward said disorder.
  • the immune-related disorder may be any one of autoimmune disease, non-alcoholic steatohepatitis, fatty liver, metabolic syndrome and any disorder associated therewith, infectious disease, and proliferative disorder.
  • the composition of the disclosure may be used for treating acute complication, or for preventing the development or recurrence of these complications.
  • the combined composition of the disclosure leads to modulation of the Th1/Th2, Tr1/Th3 cell balance toward an anti-inflammatory Th2, Tr1/Th3 immune response thereby inhibiting an immune response specifically directed toward said disorder.
  • such composition may be applicable in the treatment of an autoimmune disease.
  • the combined composition of the disclosure may lead to modulation of the Th1/Th2, Tr1/Th3 cell balance toward a pro-inflammatory Th1/Th2 immune response thereby enhancing an immune response specifically directed toward said disorder.
  • such composition may be applicable in the treatment of infectious diseases, and proliferative disorders.
  • composition of the disclosure may be administerable orally or by inhalation as an aerosol, or via intravenous, intramuscular, subcutaneous, intraperitoneal, perenteral, transdermal, intravaginal, intranasal, mucosal, sublingual, topical, rectal or subcutaneous, or any combination thereof.
  • Tolerance has been defined as a lack of response to self, or any mechanism by which a potentially injurious immune response is prevented, suppressed, or shifted to a noninjurious class of immune response.
  • tolerance is related to productive self-recognition, rather than blindness of the immune system to its own components.
  • the present inventors have demonstrated that exposure to disease-associated antigens, whether self-antigens or not, can activate some parts of the immune system while suppressing unwanted immunity in an antigen-specific manner.
  • oral antigen administration on one hand activates specific subsets of cells, suppressing specific cells and alleviating unwanted autoimmunity, and on the other hand promotes anti-viral or anti-tumor-associated antigen immune responses.
  • the balance between different types of signals/cells that are promoted in the systemic immune system will determine the final immunological effect.
  • Oral tolerance is a natural immunologic process driven by the presence of an exogenous antigen that is thought to have evolved to treat external agents that gain access to the body via a natural route and then become part of the self.
  • antigen-specific therapy seems an attractive approach for immunotherapy toward antigens present in the gut mucosa, where they can be dealt with in a noninjurious or noninflammatory immunologic environment.
  • specific immune cells may be activated and, antigen-specific therapy can serve as an immunotherapeutic chronic hepatitis, infectious agents, metabolic syndrome and other pathologic disorders discussed herein.
  • T cells The mechanisms responsible for gastrointestinal homeostasis involve a complex interplay between different types of T cells, including regulatory T cells, dendritic cells (DCs), natural killer T (NKT) cells, and the gut microenvironment.
  • DCs dendritic cells
  • NKT natural killer T
  • the follicle-associated epithelium plays key roles in antigen uptake and subsequent induction of mucosal immunity.
  • FAE M cells by targeting antigen (Ag) deliver, facilitate oral tolerance via the reduction in Ag-specific CD4+ T cells and increased levels of transforming growth factor (TGF)- ⁇ and interleukin (IL)-10-producing CD25+CD4+ T-regulatory cells (Tregs) in both systemic and mucosal lymphoid tissues.
  • TGF transforming growth factor
  • IL interleukin
  • Intestinal DCs are key regulators of pathogenic immunity, oral tolerance, and intestinal inflammation.
  • the relevant DCs may be in the PP, MLNs, or LP of the villus mucosa. All of these tissues contain a number of distinctive DC subsets, including some that can preferentially induce the differentiation of Tregs.
  • NKT cells are a unique lineage of T cells that share properties with both NK cells and memory T cells. This subset of lymphocytes may be either CD4+ or double negative and is CD1d reactive. These cells are unique in their invariant V ⁇ 14-J ⁇ 18 TCR ⁇ -chain, and their T-cell receptor (TCR) ⁇ -chain is biased toward V ⁇ 8.2, V ⁇ 2, and V ⁇ 7. NKT cells are unique in their glycolipid antigen reactivity and marked cytokine production. The ability of NKT cells to generate both Th1 and Th2 responses indicates their importance as immunoregulatory cells. The use of NKT ligands induces a profound immunomodulatory effect by altering the plasticity of these cells.
  • NKT cells produce cytokines immediately after exposure to activating signals and can determine the differentiation of Tregs.
  • the liver is considered to be important for oral tolerance.
  • the liver is a site at which apoptotic CD8+ T cells accumulate during the clearance phase of peripheral immune responses.
  • the normal mouse liver contains an unusual mixture of lymphocytes, in which natural killer (NK) and natural killer T (NKT) cells are abundant and apoptotic T cells are also present. These cells are relevant for intrahepatic T-cell trapping and killing.
  • NK natural killer
  • NKT natural killer T
  • TNT natural killer T
  • TNT natural killer T
  • TNT natural killer T
  • TNT natural killer T
  • TNT natural killer T
  • T-cell trapping and killing are relevant for intrahepatic T-cell trapping and killing.
  • Continuous exposure of diverse liver cell types to LPS derived from intestinal bacteria is thought to promote expression of cytokines, antigen-presenting molecules, and costimulatory signals that impose T-cell inactivation.
  • Other possible explanations for the tolerogenic environment in the liver involve clonal deletion, specific antigen presentation by endo
  • liver-derived DCs are inherently tolerogenic when compared with skin DCs, produce IL-10, and express low levels of costimulatory molecules. Local secretion of IL-10 and TGF- ⁇ by Kupffer cells and hepatocytes can skew DC function toward the generation of regulatory as opposed to effector pathways.
  • LSECs Liver sinusoidal endothelial cells
  • LSECs Liver sinusoidal endothelial cells
  • the outcome of antigen presentation by LSECs is usually tolerance, with apoptosis of CD8+ T cells and secretion of IL-4 and IL-10 by CD4+ T cells.
  • Activated T cells are also trapped by intercellular adhesion molecule 1 (ICAM-1)-dependent mechanisms within the sinusoids as a mechanism for regulating apoptotic pathways during control of systemic CD8 responses.
  • IAM-1 intercellular adhesion molecule 1
  • Hepatocytes themselves can function as APCs to activate naive T cells.
  • Peripheral Tregs are generated by activation of naive T cells by immature DCs or in the presence of IL-10 and TGF- ⁇ , both of which are present in the liver environment.
  • Tregs are important in the gut-liver immune axis.
  • CD4+CD25+ Tregs suppress the activation of CD4+ T cells by LSECs, Kupffer cells, or hepatocytes. Because this process can be overcome by TLR4 activation, the interaction among Tregs, pathogens, and other liver cells determines the outcome of immune activation in the liver.
  • Tregs can curb unwanted immune responses and regulate responses to the microflora and can play a role in a number of chronic inflammatory diseases of the gut. Tregs can prevent detrimental inflammatory responses against commensal organisms in the lower gut, thus guarding against inflammatory bowel diseases.
  • T lymphocytes have been suggested to exhibit regulatory functions, including natural Tregs, induced Tregs, Tr1, and Th3 cells. These cells may be activated by cytokines, and their inductive phase may be antigen driven. Most CD4+ regulatory T cells (Tr1, Th3, and CD4+CD25+) are thought to interact with dendritic cells. Other subsets of Tregs, such as CD8+ TrE cells, may recognize antigens that are presented by intestinal epithelial cells.
  • CD4+CD25+ Tregs are considered to be instrumental in regulating immune responses in the mucosa.
  • TGF- ⁇ has emerged as one of the most important cytokines produced in the gut, and its interaction with CD4+CD25+ Tregs is key in maintaining a balance between T-cell immunity and tolerance.
  • Expression of a stable form of ⁇ -catenin in CD4+CD25+ Tregs results in a marked enhancement of the survival of these cells.
  • the number of Tregs necessary for protection against inflammatory bowel disease could be substantially reduced when stable ⁇ -catenin-expressing CD4+CD25+ Tregs are used.
  • IL-35 is an inhibitory cytokine produced by Treg cells and is required for maximal suppressive activity. As discussed below, the present inventors have demonstrated modulation of CD4+ CD25+ Treg cells with compositions according to the present invention,
  • Foxp3+ Tregs are important for the establishment and maintenance of mucosal tolerance. Cytokine deprivation-induced apoptosis is a prominent mechanism by which Tregs inhibit effector TCR. As such, CD4+CD25+Foxp3+ Tregs induce apoptosis in effector CD4+ T cells.
  • TGF- ⁇ secretion by Th3 or other Treg cells is considered to be a key factor in oral tolerance.
  • TGF- ⁇ -producing cells are crucial for oral tolerance and may be master regulators of most of the mechanisms triggered by antigen feeding.
  • Latency-associated peptide (LAP) is the amino-terminal domain of the TGF- ⁇ precursor peptide, and remains noncovalently associated with the TGF- ⁇ peptide after cleavage and forms the latent complex.
  • LAP Latency-associated peptide
  • the presence of membrane-bound TGF- ⁇ or LAP on the surface of Tregs has linked TGF- ⁇ with the suppressive function of Tregs.
  • TGF- ⁇ -secreting Th3 cells and CD8+ regulatory cells have been associated with oral tolerance and are dependent on TGF- ⁇ .
  • the present inventors have demonstrated modulation of LAP+ and LAP- Treg cells with compositions according to the present invention,
  • TGF- ⁇ containing LAP A membrane-bound form of TGF- ⁇ containing LAP has been described.
  • LAP+CD4+ cells mediate suppression in the gut via a TGF- ⁇ -dependent mechanism.
  • the present inventors have shown that TGF- ⁇ -dependent Tregs that express surface LAP are induced/promoted by oral administration of anti-LPS antibodies.
  • TGF- ⁇ may induce the differentiation of IL-10-producing cells, indicating that cross-talk between different cytokine-producing Tregs may exist in oral tolerance induction, for example inducing CD4+CD25-LAP+ Tregs, which suppress autoimmunity.
  • IECs Intestinal epithelial cells
  • T cells activated by IECs are suppressive in function, whereas IECs can induce the proliferation of a small fraction of CD8+ peripheral T cells.
  • the CD8+CD28- subset of IEC-activated CD8+ T cells expresses CD101 and CD103, interacts with IECs through gp180, and possesses a regulatory function.
  • CD8+ T cells with regulatory activity are present in the LP of normal, healthy individuals, but not in patients with inflammatory bowel disease (IBD), indicating that these cells play an active role in mucosal tolerance.
  • IBD inflammatory bowel disease
  • Antigen-cross-presentation or the possibility that molecules presented by professional APCs can leak into the major histocompatibility complex class I (MHC-I) pathway and are presented to CD8+ T cells, is a possible mechanism.
  • MHC-I major histocompatibility complex class I
  • cross-priming of CD8+ by APCs associated with CD4+ T-cell activation may be a mechanism responsible for suppression.
  • CD8+ T cells play a regulatory role via secretion of TGF- ⁇ .
  • Antigen-primed CD8+ T-cell populations produce IL-4 or IL-10, and may be associated with tolerance induction.
  • the present disclosure provides a composition comprising a mammalian anti-LPS enriched colostrum-derived immunoglobulin preparation for inducing CD4+ CD25+ T cells in the liver, inducing CD4+ CD25+ LAP- T cells in the liver, inducing CD45+ LAP+ T cells in the liver, inducing CD3+ LAP+ T cells in the liver, inducing CD45+ LAP+ T cells in the spleen, inducing CD8+ LAP+ T cells in the spleen, inducing CD3+ LAP+ T cells in the spleen, inducing CD8+ CD25+ T cells in the spleen, inducing CD4+ CD25+ T cells in adipose tissue, inducing CD3+ LAP+ T cells in adipose tissue, inducing CD4+ CD25+ T cells in stromal vascular cells, inducing CD4+ CD25+ LAP+ T cells in stromal vascular cells, inducing CD4
  • Adipocytes are the cells that primarily compose adipose tissue, specialized in storing energy as fat.
  • adipose tissue white adipose tissue (WAT) and brown adipose tissue (BAT), which are also known as white fat and brown fat, respectively, and comprise the two types of fat cells.
  • WAT white adipose tissue
  • BAT brown adipose tissue
  • White fat cells or monovacuolar cells contain a large lipid droplet surrounded by a layer of cytoplasm. The nucleus is flattened and located on the periphery.
  • a typical fat cell is 0.1mm in diameter with some being twice that size and others half that size.
  • the fat stored is in a semi- liquid state, and is composed primarily of triglycerides and cholesteryl ester.
  • White fat cells secrete resistin, adiponectin, and leptin.
  • Brown fat cells or pluri vacuolar cells are polygonal in shape. Unlike white fat cells, these cells have considerable cytoplasm, with lipid droplets scattered throughout. The nucleus is round, and, although eccentrically located, it is not in the periphery of the cell. The brown color comes from the large quantity of mitochondria.
  • the compositions of the invention significantly decreased the serum levels of triglycerides, ALT, AST and glucose. Therefore, according to one embodiment, the pharmaceutical composition of the invention leads to at least one of a decrease in the serum levels of cholesterol, triglycerides, ALT, AST and glucose and a decrease in insulin resistance in a subject suffering of a liver disorder or an immune -related disorder, for example, Metabolic syndrome. Wherein indicated decease, reduction, inhibition, it is meant that the composition of the invention leads to a reduction of about 5% to 99% of the serum level of any one of triglycerides, ALT, AST and Glucose, in a subject suffering of an-immune- related disorder.
  • such reduction may be a reduction of about, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% and over 99%, as compared to the levels prior to the treatment, or the levels of untreated control.
  • increase, elevation, enhancement, induction it is meant that the composition of the invention leads to induction, or increase of about 5% to 99%. More specifically, such increase may be an.
  • composition of the disclosure may be used for preventing and/or treating autoimmune disease for example, Metabolic Syndrome or any of the conditions comprising the same, any condition associated with, caused by, linked to or believed to have an impact on metabolic syndrome, for example, at least one of dyslipoproteinemia (hypertriglyceridemia, hypercholesterolemia, low HDL-cholesterol), obesity, NIDDM (non-insulin dependent diabetes mellitus), IGT (impaired glucose tolerance), blood coagulability, blood fibrinolysis defects and hypertension.
  • dyslipoproteinemia hypertriglyceridemia, hypercholesterolemia, low HDL-cholesterol
  • obesity non-insulin dependent diabetes mellitus
  • IGT impaired glucose tolerance
  • blood coagulability blood fibrinolysis defects and hypertension.
  • composition of the disclosure is intended for the treatment of dyslipoproteinemia, which may include hypertriglyceridemia, hypercholesterolemia and low HDL-cholesterol, obesity, NIDDM (non- insulin dependent diabetes mellitus type 2), IGT (impaired glucose tolerance), blood coagulability, blood fibronolysis defects and hypertension.
  • dyslipoproteinemia may include hypertriglyceridemia, hypercholesterolemia and low HDL-cholesterol, obesity, NIDDM (non- insulin dependent diabetes mellitus type 2), IGT (impaired glucose tolerance), blood coagulability, blood fibronolysis defects and hypertension.
  • the immunomodulatory composition of the disclosure may be used for treating diabetes, particularly, Type 2 diabetes.
  • Diabetes mellitus often simply diabetes, is a syndrome characterized by disordered metabolism and inappropriately high, blood sugar (hyperglycaemia) resulting from either low levels of the hormone insulin or from abnormal resistance to insulin's effects coupled with inadequate levels of insulin secretion to compensate.
  • the characteristic symptoms are excessive urine production (polyuria), excessive thirst and increased fluid intake (polydipsia), and blurred vision. These symptoms are likely absent if the blood sugar is only mildly elevated.
  • Type 1 diabetes is usually due to autoimmune destruction of the pancreatic beta cells.
  • Type 2 diabetes is characterized by insulin resistance in target tissues, this causes a need for abnormally high amounts of insulin and diabetes develops when the beta cells cannot meet this demand.
  • Gestational diabetes is similar to type 2 diabetes in that it involves insulin resistance, hormones in pregnancy may cause insulin resistance in women genetically predisposed to developing this condition.
  • Acute complication of diabetes may occur if the disease is not adequately controlled.
  • Serious long-term complications include cardiovascular disease (doubled risk), chronic renal failure, retinal damage (which can lead to blindness), nerve damage (of several kinds), and microvascular damage, which may cause impotence and poor healing. Poor healing of wounds, particularly of the feet, can lead to gangrene, which may require amputation.
  • the immunomodulatory composition of the disclosure may be used for the treatment of Type 1 diabetes.
  • Type 1 diabetes mellitus is characterized by loss of the insulin-producing beta cells of the islets of Langerhans in the pancreas, leading to a deficiency of insulin. The main cause of this beta cell loss is. a T-cell mediated autoimmune attack.
  • the pharmaceutical composition of the disclosure may be used for the treatment of an autoimmune disorder.
  • autoimmune disorders include, but are not limited to, Alopecia Areata, Lupus, Ankylosing Spondylitis, Meniere's Disease, Antiphospholipid Syndrome, Mixed Connective Tissue Disease, Autoimmune Addison's Disease, Multiple Sclerosis, Autoimmune Hemolytic Anemia, Myasthenia Gravis, Autoimmune Hepatitis, Pemphigus Vulgaris, Behcet's Disease, Pernicious Anemia, Bullous Pemphigoid, Polyarthritis Nodosa, Cardiomyopathy, Polychondritis, Celiac Sprue-Dermatitis, Polyglandular Syndromes, Chronic Fatigue Syndrome (CFIDS), Polymyalgia Kheumatica, Chronic Inflammatory Demyelinating, Polymyositis and Dermatomyositis, Chronic Inflammatory Polyneuropathy, Primary Agammaglobulinemia, Chur
  • compositions described herein can be administered to a subject to treat or prevent disorders associated with an abnormal or unwanted immune response associated with cell, tissue or organ transplantation, e.g., renal, hepatic, and cardiac transplantation, e.g., graft versus host disease (GVHD), or to prevent allograft rejection.
  • disorders associated with an abnormal or unwanted immune response associated with cell, tissue or organ transplantation e.g., renal, hepatic, and cardiac transplantation, e.g., graft versus host disease (GVHD), or to prevent allograft rejection.
  • GVHD graft versus host disease
  • an autoimmune disease treated by the composition of the disclosure may be any one of rheumatoid arthritis, type 1 diabetes, type 2 diabetes, artherosclerosis, asthma, acute and chronic graft versus host disease, systemic lupus erythmatosus, scleroderma, multiple sclerosis, inflammatory bowel disease, psoriasis, uvietis, thyroiditis and immune mediated hepatitis.
  • the composition of the disclosure may be used for the treatment of MS.
  • MS Multiple Sclerosis
  • MS is typically characterized clinically by recurrent or chronically progressive necrologic dysfunction, caused by lesions in the CNS.
  • the lesions include multiple areas of demyelination affecting the brain, optic nerves, and spinal cord.
  • the underlying etiology is uncertain, but MS is widely believed to be at least partly an autoimmune or immune-mediated disease.
  • the disclosure includes compositions and methods of treating, delaying or preventing the onset of MS, by orally or mucosally administering the colostrum- derived immunoglobulin preparation of the disclosure. Included are methods wherein a subject who has or is at risk of having MS is orally administered with the composition of the disclosure.
  • the composition of the disclosure may be used for the treatment of RA.
  • Rheumatoid arthritis is the most common chronic inflammatory arthritis and affects about 1% of adults, it is two to three times more prevalent in women than in men. RA may begin as early as infancy, but onset typically occurs in the fifth or sixth decade.
  • Diagnosis may be made according to the American Rheumatism Association Criteria for the so Classification of Rheumatoid Arthritis.
  • a therapeutically effective amount will cause an improvement in one or more of the following: the number of inflamed joints, the extent of swelling, and the range of joint motion.
  • Laboratory measurements e.g., ESR and hematocrit value
  • assessments of subjective features e.g., pain and morning stiffness
  • the disclosure also includes methods of treating autoimmune arthritis, e.g., RA, in a subject by administering to the subject a therapeutically effective amount of composition of the disclosure comprising colostrum-derived immunoglobulin preparations.
  • compositions of the disclosure described herein can also be used to treat or prevent graft rejection in a transplant recipient.
  • the compositions can be used in a wide variety of tissue and organ transplant procedures, e.g., the compositions can be used to induce central tolerance in a recipient of a graft of cells, e. g., stem cells such as bone marrow and/or of a tissue or organ such as pancreatic islets, liver, kidney, heart, lung, skin, muscle, neuronal tissue, stomach, and intestines.
  • the new methods can be applied in treatments of diseases or conditions that entail cell, tissue or organ transplantation (e.g., liver transplantation to treat hypercholesterolemia, transplantation of muscle cells to treat muscular dystrophy, or transplantation of neuronal tissue to treat Huntington's disease or Parkinson's disease).
  • diseases or conditions that entail cell, tissue or organ transplantation (e.g., liver transplantation to treat hypercholesterolemia, transplantation of muscle cells to treat muscular dystrophy, or transplantation of neuronal tissue to treat Huntington's disease or Parkinson's disease).
  • the composition of the disclosure may modulate the Th1/Th2, Th3 balance towards an anti-Th2, Trl/Th3 response in a subject suffering from IBD. Therefore, according to this embodiment, the composition of the disclosure is intended for treating IBD.
  • IBD Inflammatory bowel diseases
  • Th1-pro-inflammatory, Th2-anti-inflammatory subtypes of immune responses are common gastrointestinal disorders that can be perceived as being the result of a dysbalance between Th1-pro- inflammatory, and Th2-anti-inflammatory subtypes of immune responses.
  • T cell-mediated immunity including coetaneous anergy and diminished responsiveness to T cell stimuli, have also been described in these patients.
  • changes in mucosal cell mediated immunity were identified, including increased concentrations of mucosal IgG cells and changes in T cells subsets, suggesting antigen stimulation.
  • the composition of the disclosure may be used for the treatment of atherosclerosis.
  • Atherosclerosis is a slowly progressive disease characterized by the accumulation of cholesterol within the arterial wall.
  • the atherosclerotic process begins when LDL-C becomes trapped within the vascular wall. Oxidation of the LDL-C results in the bonding of monocytes to the endothelial cells lining the vessel wall. These monocytes are activated and migrate into the endothelial space where they are transformed into macrophages, leading to further oxidation of LDL-C.
  • the oxidized LDL-C is taken up through the scavenger receptor on the macrophage leading the formation of foam cells.
  • a fibrous cap is generated through the proliferation and migration of arterial smooth muscle cells, thus creating an atherosclerotic plaque.
  • Lipids depositing in atherosclerotic legions are derived primarily from plasma apo B containing lipoproteins. These include chylomicrons, LDL-C, IDL, and VLDL. This accumulation forms bulky plaques that inhibit the flow of blood until a clot eventually forms, obstructing an artery and causing a heart attack or stroke.
  • the immunoglobulin preparation used by the composition of the disclosure may recognize and bind at least one antigen specific for the treated disorder and may modulates immune-regulatory cells, specifically, regulatory T cells. Such modulation may results for example, in modulation of the Th1/Th2 cell balance toward a pro-inflammatory ThI immune response thereby activating an immune response specifically directed toward said disorder.
  • the pro -inflammatory effect of the immunomodulatory composition of the disclosure may be achieved by activation or promotion of specific subsets of regulatory cells, antigen presenting cells or any type of cell-cell contact via direct or indirect activation, of cytokines, and/or chemokines.
  • modulation of the Th1/Th2, Th3 balance towards a pro-inflammatory Th1 response may be particularly applicable in immune related disorders having an undesired unbalanced anti-inflammatory Th2, Tr1/Th3 response, for example, a malignant and non-malignant proliferative disorder, infectious disease, genetic disease and neurodegenerative disorders.
  • the present disclosure provides a use of an anti-LPS enriched immunoglobulin preparation in the manufacture of a medicament for the treatment and/or prophylaxis of a pathologic disorder.
  • the anti-LPS enriched immunoglobulin preparation may be derived from colostrum or from avian eggs.
  • the pathologic disorder is acute or chronic liver disease, cirrhosis or any disease or complication associated therewith.
  • the acute or chronic liver disease, cirrhosis and any disease or complication associated therewith is selected from the group consisting of hepatic encephalopathy, spontaneous bacterial peritonitis (SBP), ascites, bleeding varices, cirrhosis associated hyperdynamic circulation, hepatorenal syndrome, hepatopulmonary syndrome, portopulmonary hypertension, variceal bleeding, adrenal insufficiency and altered level of consciousness.
  • SBP spontaneous bacterial peritonitis
  • the medicament is for the treatment and/or prophylaxis of liver damage.
  • the pathologic disorder is an immune-related disorder selected from the group consisting of autoimmune disease, non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith, infectious disease, and proliferative disorder.
  • the pathologic disorder is selected from the group consisting of secondary peritonitis and infection after surgery, hepatic cardiomyopathy and hypotension, hepatoadrenal syndrome, hepatocellular carcinoma, Alzheimer's disease, any type of memory loss, any type of dementia, attention deficit disorders (ADHA), any type of learning disability, effect of alcohol or drugs on the brain, any type of immune mediated disease including asthma, and peritonitis.
  • the medicament may further comprise an immunoglobulin preparation comprising immunoglobulins that recognize and bind at least one antigen specific for said pathologic disorder.
  • the further immunoglobulin preparation may be derived from colostrum.or from avian eggs.
  • the medicament modulates regulatory T cells leading to modulation of the Th1/Th2, Tr1/Th3 cell balance toward an anti-inflammatory Th2, Tr1/Th3 immune response or a pro-inflammatory Th1 immune response thereby inhibiting or activating an immune response specifically directed toward said disorder.
  • the medicament modulates the Th1/Th2, Tr1/Th3 cell balance toward an anti-inflammatory Th2, Tr1/Th3 immune response thereby inhibiting an immune response specifically directed toward said disorder
  • said composition is for the treatment of any one of an autoimmune disease, non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith selected from diabetes type 2, insulin resistance, obesity and overweight.
  • the medicament is for the treatment and/or prophylaxis of metabolic syndrome or non-alcoholic steatohepatitis or both, the treatment and/or prophylaxis of diabetes, the treatment impaired glucose tolerance, such as decreasing glucose tolerance, decreasing serum insulin levels, decreasing hepatic triglyceride levels, or decreasing cholesterol levels.
  • the medicament modulates the Th1/Th2, Tr1/Th3 cell balance toward a pro-inflammatory Th1/Th2 immune response thereby enhancing an immune response specifically directed toward said disorder, and wherein said composition is for the treatment of infectious diseases, and proliferative disorders,
  • the medicament may further comprise a therapeutic agent, carrier or adjuvant and/or non-hyperimmune colostrum.
  • the medicament is formulated for administration orally, by inhalation as an aerosol, or by parenteral, intravaginal, intranasal, mucosal, sublingual, topical, or rectal administration, or any combination thereof.
  • the immunoglobulin preparation or any fractions thereof recognizes and binds LPS or any fragments thereof.
  • composition reduces or inhibits mucosal microbial translocation. In another embodiment the composition reduces or inhibits mucosal microbial translocation and thereby modulates immune activation.
  • the present disclosure provided a use of a mammalian anti-LPS enriched colostrum-derived immunoglobulin preparation in the manufacture of a medicament for modulating immune tolerance in a subject, or in another embodiment, a medicament for modulating oral tolerance in a subject.
  • the present disclosure provides the use of a mammalian anti-LPS enriched colostrum-derived immunoglobulin preparation in the manufacture of a medicament for inducing CD4+ CD25+ T cells in the liver, inducing CD4+ CD25+ LAP- T cells in the liver, inducing CD45+ LAP+ T cells in the liver, inducing CD3+ LAP+ T cells in the liver, inducing CD45+ LAP+ T cells in the spleen, inducing CD8+ LAP+ T cells in the spleen, inducing CD3+ LAP+ T cells in the spleen, inducing CD8+ CD25+ T cells in the spleen, inducing CD4+ CD25+ T cells in adipose tissue, inducing CD3+ LAP+ T cells in adipose tissue, inducing CD4+ CD25+ T cells in stromal vascular cells, inducing CD4+ CD25+ LAP+ T cells in stromal
  • the anti-LPS enriched immunoglobulin preparation may be derived from colostrum or from avian eggs.
  • the present disclosure provides a method for the treatment and/or prophylaxis of a pathologic disorder comprising the step of administering to a subject in need thereof a therapeutically effective amount of a composition comprising an anti-LPS enriched immunoglobulin preparation.
  • the anti-LPS enriched immunoglobulin preparation may be derived from colostrum or from avian eggs.
  • the pathologic disorder is acute or chronic liver disease, cirrhosis or any disease or complication associated therewith.
  • the acute or chronic liver disease, cirrhosis and any disease or complication associated therewith is selected from the group consisting of hepatic encephalopathy, spontaneous bacterial peritonitis (SBP), ascites, bleeding varices, cirrhosis associated hyperdynamic circulation, hepatorenal syndrome, hepatopulmonary syndrome, portopulmonary hypertension, variceal bleeding, adrenal insufficiency and altered level of consciousness.
  • SBP spontaneous bacterial peritonitis
  • the pathologic disorder is liver damage.
  • the pathologic disorder is an immune-related disorder selected from the group consisting of autoimmune disease, non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith, infectious disease, and proliferative disorder.
  • the pathologic disorder is selected from the group consisting of secondary peritonitis and infection after surgery, hepatic cardiomyopathy and hypotension, hepatoadrenal syndrome, hepatocellular carcinoma, Alzheimer's disease, any type of memory loss, any type of dementia, attention deficit disorders (ADHA), any type of learning disability, effect of alcohol or drugs on the brain, any type of immune mediated disease including asthma, and peritonitis.
  • the composition further comprises an immunoglobulin preparation comprising immunoglobulins that recognize and bind at least one antigen specific for said pathologic disorder.
  • the further immunoglobulin preparation may be derived from colostrum.or from avian eggs.
  • the composition modulates regulatory T cells leading to modulation of the Th1/Th2, Tr1/Th3 cell balance toward an anti-inflammatory Th2, Tr1/Th3 immune response or a pro-inflammatory Th1 immune response thereby inhibiting or activating an immune response specifically directed toward said disorder.
  • the composition modulates the Th1/Th2, Tr1/Th3 cell balance toward an anti-inflammatory Th2, Tr1/Th3 immune response thereby inhibiting an immune response specifically directed toward said disorder, and wherein said composition is for the treatment of any one of an autoimmune disease, non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith selected from diabetes type 2, insulin resistance, obesity and overweight.
  • the pathologic disorder is metabolic syndrome or non-alcoholic steatohepatitis or both.
  • the pathologic disorder is diabetes. In another embodiment, the pathologic disorder is impaired glucose tolerance.
  • the method decreases glucose tolerance, decreases serum insulin levels, decreases hepatic triglyceride levels, or decreases cholesterol levels.
  • the method modulates the Th1/Th2, Tr1/Th3 cell balance toward a pro-inflammatory Th1/Th2 immune response thereby enhancing an immune response specifically directed toward said disorder, and wherein said composition is for the treatment of infectious diseases, and proliferative disorders,
  • composition further comprises non-hyperimmune colostrum and/or a therapeutic agent, carrier or adjuvant.
  • composition may be administered orally, by inhalation as an aerosol, or by parenteral, intravaginal, intranasal, mucosal, sublingual, topical, or rectal administration, or any combination thereof.
  • the immunoglobulin preparation or any fractions thereof recognizes and binds LPS or any fragments thereof.
  • the method reduces or inhibits mucosal microbial translocation. In another embodiment, the method reduces or inhibits mucosal microbial translocation and thereby modulates immune activation.
  • the present disclosure provides a method for modulating immune tolerance in a subject comprising the step of administering to a subject in need thereof a therapeutically effective amount of a composition comprising a mammalian anti-LPS enriched colostrum-derived immunoglobulin preparation.
  • the method may be for modulating oral tolerance.
  • a method for inducing CD4+ CD25+ T cells in the liver of a subject comprising the step of administering to a subject in need thereof a therapeutically effective amount of a composition comprising a mammalian anti-LPS enriched colostrum-derived immunoglobulin preparation.
  • the method may be for inducing CD4+ CD25+ LAP- T cells in the liver, CD45+ LAP+ T cells in the liver, inducing CD3+ LAP+ T cells in the liver, inducing CD45+ LAP+ T cells in the spleen, inducing CD8+ LAP+ T cells in the spleen, inducing CD3+ LAP+ T cells in the spleen, inducing CD8+ CD25+ T cells in the spleen, inducing CD4+ CD25+ T cells in adipose tissue, inducing CD3+ LAP+ T cells in adipose tissue, inducing CD4+ CD25+ T cells in stromal vascular cells, inducing CD4+ CD25+ LAP+ T cells in stromal vascular cells, decreasing CD3+ NK1.1+ cells in the liver, decreasing CD25+ LAP- T cells in the liver, decreasing CD25+ LAP+ T cells in the liver, inducing CD4+ CD25+
  • the present disclosure provides a method for the treatment and/or prophylaxis of a pathologic disorder.
  • the method of the disclosure comprises the step of administering to a subject in need thereof a therapeutically effective amount of a mammalian colostrum-derived anti-LPS enriched immunoglobulin preparation or of a composition comprising the same. It should be noted that the immunoglobulin preparation or any fractions thereof recognizes and binds LPS and any fragments thereof.
  • the method of the disclosure comprises the step of administering a combined composition of anti-LPS enriched immunoglobulin preparation of the disclosure with at least one immunoglobulin preparation comprising immunoglobulins recognizing at least one antigen specific for said disorder, thereby activating or inhibiting an immune response specifically directed toward said disorder.
  • the colostrum-derived, milk or milk product/s-derived anti-LPS enriched immunoglobulin preparation or any fragment or mixture, combination, or any composition thereof, used by the method of the disclosure comprises a monomeric, dimeric and multimeric immunoglobulin selected from the group consisting of IgG, IgA and IgM and any fragments thereof, preparations, mixtures and compositions thereof. More specifically, the immunoglobulin preparation used by the method of the disclosure may specifically comprise IgG, particularly, IgG1 and/or IgG2 and any fragments thereof. Alternatively or additionally, the immunoglobulin preparation used by the method of the disclosure may specifically comprise secretory dimeric IgA.
  • the method of the disclosure may use a composition or combined composition comprising colostrum-derived anti-LPS enriched immunoglobulin preparation.
  • Such composition optionally further comprises colostrum component/s, preferably, alarmins, defenensins, colostrinin, or any glycolipids, carbohydrates or any preparations, mixtures and combinations thereof, or any other adjuvant/s.
  • colostrum component/s preferably, alarmins, defenensins, colostrinin, or any glycolipids, carbohydrates or any preparations, mixtures and combinations thereof, or any other adjuvant/s.
  • the present disclosure further provides the use of colostrum or any colostrum-derived preparations for enhancing an immunomodulatory effect of an immunomodulatory therapeutic agent.
  • the composition or combined composition used by the method of the disclosure may comprise any additional adjuvant.
  • Appropriate adjuvants therefore may be any antigen, antibody, glycosphingolipids, proteins, cytokines, adhesion molecules, and component that can activate or alter the function of antigen presenting cell or of any other cell related to the immune system in a direct and indirect manner.
  • the anti-LPS enriched immunoglobulin preparation or any other immunoglobulin preparation used by the disclosure may be obtained from a mammal, preferably a cow, immunized with LPS and optionally, in addition, with at least one antigen or a mixture of at least two antigens specific for a disorder to be treated.
  • the method of the disclosure comprises the step of administering to said subject a therapeutically effective amount of a mammalian anti-lipopolysaccharide (LPS) colostrum-derived immunoglobulin preparation or any composition comprising the same.
  • LPS mammalian anti-lipopolysaccharide
  • colostrum-derived immunoglobulin preparation or any composition comprising the same. It should be noted that such method may be particularly applicable for the treatment, prevention and prophylaxis of acute or chronic liver disease, cirrhosis and any disease or complication associated therewith.
  • liver disease More specifically, acute or chronic liver disease, cirrhosis and any disease or complication associated therewith is at least one of hepatic encephalopathy, spontaneous bacterial peritonitis (SBP), ascitess, cirrhosis associated hyperdynamic circulation, hepatorenal syndrome, hepatopulmonary syndrome, portopulmonary hypertension, variceal bleeding, adrenal insufficiency and altered level of consciousness.
  • SBP spontaneous bacterial peritonitis
  • ascitess cirrhosis associated hyperdynamic circulation
  • hepatorenal syndrome hepatopulmonary syndrome
  • portopulmonary hypertension variceal bleeding
  • adrenal insufficiency and altered level of consciousness.
  • the disclosure provides a method for treating immune-related disorders.
  • the method of the disclosure comprises the step of administering to said subject a therapeutically effective amount of a combination of anti-LPS enriched immunoglobulin preparation with at least one colostrum-derived immunoglobulin preparation comprising immunoglobulins that recognize and bind at least one antigen specific for said pathologic disorder, or of a combined composition comprising the same and optionally an additional therapeutic agent or any carrier and adjuvant.
  • the combination used by the invention modulates regulatory T cells leading to modulation of the Th1/Th2, Tr1/Th3 cell balance toward an anti-inflammatory Th2, Tr1/Th3 immune response or a pro-inflammatory Th1 immune response thereby inhibiting or activating an immune response specifically directed toward said disorder.
  • the method of the disclosure may be particularly applicable or treating an immune-related disorder, for example, autoimmune disease, non-alcoholic steatohepatitis, fatty liver, metabolic syndrome and any disorder associated therewith, infectious disease, and proliferative disorder.
  • an immune-related disorder for example, autoimmune disease, non-alcoholic steatohepatitis, fatty liver, metabolic syndrome and any disorder associated therewith, infectious disease, and proliferative disorder.
  • the present disclosure provides a method of treating impaired glucose tolerance.
  • the present disclosure provides a method of decreasing glucose tolerance.
  • the present disclosure provides a method of decreasing serum insulin levels
  • the present disclosure provides a method of decreasing hepatic triglyceride levels.
  • the present disclosure provides a method of decreasing cholesterol levels.
  • the method of the disclosure is for treatment of acute complications, for preventing the development and/or the recurrence of these complications.
  • the combined composition used by the method of the disclosure leads to modulation of the Th1/Th2, Tr1/Th3 cell balance toward an anti-inflammatory Th2, Tr1/Th3 immune response thereby inhibiting an immune response specifically directed toward said disorder.
  • such composition may be applicable in the treatment of any one of an autoimmune disease, non-alcoholic steatohepatitis, fatty liver, atherosclerosis, metabolic syndrome and any disorder associated therewith for example, diabetes type 2, insulin resistance, obesity and overweight.
  • the combined composition used by the method of the disclosure may lead to modulation of the Th1/Th2, Tr1/Th3 cell balance toward a pro-inflammatory Th1/Th2 immune response thereby enhancing an immune response specifically directed toward said disorder.
  • such composition may be applicable in the treatment of infectious disease, and proliferative disorder.
  • the method of the disclosure may be specifically applicable for treating viral disease including HCV, HBV, CMV, and EBV.
  • the anti-LPS-enriched immunoglobulin preparation is to be administered orally or by inhalation as an aerosol, or by intravenous, intramuscular, subcutaneous, intraperitoneal, parenteral, transdermal, intravaginal, intranasal, mucosal, sublingual, topical, rectal or subcutaneous administration, or any combination thereof.
  • the method of the invention is specifically suitable for the treatment of a mammalian subject.
  • mammalian for purposes of treatment refers to any animal classified as a mammal including, human, research animals, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, horses, cats, cows, etc.
  • said mammalian subject is a human subject.
  • Treatment refers to therapeutic treatment. Those in need of treatment are mammalian subjects suffering from an immune-related disease.
  • patient or “subject in need” is meant any mammal for which administration of the immuno-modulatory composition of the invention is desired, in order to prevent, overcome or slow down such infliction.
  • the terms “effective amount” or “sufficient amount” mean an amount necessary to achieve a selected result.
  • the “effective treatment amount” is determined by the severity of the disease in conjunction with the preventive or therapeutic objectives, the route of administration and the patient's general condition (age, sex, weight and other considerations known to the attending physician).
  • the dosage of needed to achieve a therapeutic effect will depend not only on such factors as the age, weight and sex of the patient and mode of administration, but also on the degree of disease progression and the potency of the particular derivative being utilized for the particular disorder of disease concerned.
  • Such method may comprise the administration of a prophylactically effective amount of the composition of the invention or of the active ingredients comprised within such composition, to a person at risk of developing a disease.
  • prophylactically effective amount is intended to mean that amount of a pharmaceutical combined composition that will prevent or reduce the risk of occurrence of the biological or medical event that is sought to be prevented in a tissue, a system, animal or human by a researcher, veterinarian, medical doctor or other clinician.
  • said therapeutic effective amount, or dosage is dependent on severity and responsiveness of the disease state to be treated, with the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved.
  • Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient. Persons of ordinary skill can easily determine optimum dosages, dosing methodologies and repetition rates. In general, dosage is calculated according to body weight, and may be given once or more daily, weekly, monthly or yearly, or even once every 2 to 20 years.
  • the present disclosure provides a method for treating a human subject with a condition selected from the group consisting of hypertension, increase in body mass index (BMI), increase in waist circumference, dyslipidemia, insulin resistance, elevated liver enzymes, and fatty liver comprising administering to the subject an effective amount of a composition comprising an anti-LPS immunoglobulin preparation.
  • a condition selected from the group consisting of hypertension, increase in body mass index (BMI), increase in waist circumference, dyslipidemia, insulin resistance, elevated liver enzymes, and fatty liver comprising administering to the subject an effective amount of a composition comprising an anti-LPS immunoglobulin preparation.
  • compositions comprising an anti-LPS immunoglobulin preparation for use in treatment and/or prophylaxis of a human subject with a condition selected from the group consisting of hypertension, increase in body mass index (BMI), increase in waist circumference, dyslipidemia, insulin resistance, elevated liver enzymes, and fatty liver.
  • BMI body mass index
  • dyslipidemia insulin resistance
  • elevated liver enzymes and fatty liver.
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for the treatment and/or prophylaxis of a human subject with a condition selected from the group consisting of hypertension, increase in body mass index (BMI), increase in waist circumference, dyslipidemia , insulin resistance, elevated liver enzymes, and fatty liver.
  • a condition selected from the group consisting of hypertension, increase in body mass index (BMI), increase in waist circumference, dyslipidemia , insulin resistance, elevated liver enzymes, and fatty liver.
  • hypertension encompasses high blood pressure, in which both the systolic and diastolic pressure levels are elevated (e.g. ⁇ 140 mmHg/ ⁇ 90 mmHg), and isolated systolic hypertension, in which only the systolic pressure is elevated, for example elevated to greater than or equal to 140 mm Hg, while the diastolic pressure is less than 90 mm Hg, and isolated diastolic hypertension, in which only the diastolic pressure is elevated, for example elevated to greater than or equal to 90 mmHg.
  • Normal blood pressure may be defined as less than 120 mmHg systolic and less than 80 mmHg diastolic.
  • a hypertensive subject is a subject with hypertension.
  • a pre-hypertensive subject is a subject with a blood pressure that is between 120 mmHg over 80 mmHg and 139 mmHg over 89 mmHg.
  • One outcome of treatment is decreasing blood pressure in a subject with high blood pressure.
  • the hypertension may characterized by a blood pressure of >120 mmHg / 80 mmHg, a blood pressure of >130 mmHg / 90 mmHg or a blood pressure of >140 mmHg / 90 mmHg.
  • Treatment of hypertension using the compositions of the present disclosure may result in a decrease in blood pressure of about 1, 2, 3, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 and over 99% relative to untreated control, or levels prior to the treatment.
  • the term "obesity” as used herein is a condition in which there is an excess of body fat.
  • An operational definition of obesity is based on the Body Mass Index (BMI), which is calculated as body weight per height in meters squared (kg/m2).
  • BMI Body Mass Index
  • "Obesity” refers to a condition whereby an otherwise healthy subject has a Body Mass Index (BMI) greater than or equal to 30 kg/m2, or a condition whereby a subject with at least one co-morbidity has a BMI greater than or equal to 27 kg/m2.
  • An "obese subject” is an otherwise healthy subject with a Body Mass Index (BMI) greater than or equal to 30 kg/m2 or a subject with at least one co-morbidity with a BMI greater than or equal to 27 kg/m2.
  • a “subject at risk of obesity” also referred to as “overweight” or “pre-obese” is an otherwise healthy subject with a BMI of 25 kg/m2 to less than 30 kg/m2 or a subject with at least one co-morbidity with a BMI of 25 kg/m2 to less than 27 kg/m2.
  • a "normal” BMI is 18.5 to 24.9 kg/m2.
  • BMI Body Mass Index
  • Exemplary measurements cutoffs for different populations are well known, for example, those in discussed in the WHO publication "Redefining obesity and its treatment", World Health Organization Western Pacific Region, International Association for the Study of Obesity, International Obesity Task Force (2000)
  • Obesity-induced or obesity-related co-morbidities include, but are not limited to, diabetes, non-insulin dependent diabetes mellitus-type 2, diabetes associated with obesity, impaired glucose tolerance, impaired fasting glucose, insulin resistance syndrome, dyslipidemia, hypertension, hypertension associated with obesity, hyperuricacidemia, gout, coronary artery disease, myocardial infarction, angina pectoris, sleep apnea syndrome, Pickwickian syndrome, fatty liver; cerebral infarction, cerebral thrombosis, transient ischemic attack, orthopedic disorders, arthritis deformans, lumbodynia, emmeniopathy, and infertility.
  • co-morbidities include: hypertension, hyperlipidemia, dyslipidemia, glucose intolerance, cardiovascular disease, sleep apnea, diabetes mellitus, and other obesity related conditions.
  • Treatment of increase in BMI or prevention of increase in BMI includes the treatment of increases in BMI, for example from normal to pre-obese, normal to obese, or pre-obese to obese.
  • the increased BMI may be a BMI of at least 25 kg/m2 to less than 30 kg/m2 or a BMI of at least 30 kg/m2.
  • the increased waist circumference may be a waist circumference of at least 102 cm in men or a waist circumference of at least 88 cm in women.
  • a 'abdominal obesity' is defined by a waist circumference greater than 102 cm (40 inches) in men or 88 cm (35 inches) in women according to NCEP ATP III.
  • Treatment of increased BMI or waist circumference using the compositions of the present disclosure may result in a decrease in BMI, or waist circumference, respectively, of about 1, 2, 3, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 and over 99% relative to untreated control, or levels prior to the treatment.
  • Methodabolic syndrome or “syndrome X” is defined here on the basis of NCEP ATP III criteria, which are the presence of three or more of the following factors: 1) increased waist circumference (>102 cm (>40 in) for men, >88 cm (>35 in) for women); 2) elevated triglycerides (>150 mg/dl); 3) low HDL cholesterol ( ⁇ 40 mg/dl in men, ⁇ 50 mg/dl in women); 4) non-optimal blood pressure (>130 mmHg systolic or ⁇ 5 mmHg diastolic); and 5) impaired fasting glucose (>110 mg/dl).
  • Dyslipidemias or disorders of lipid metabolism include various conditions characterized by abnormal concentrations of one or more lipids (i.e. cholesterol and triglycerides), and/or apolipoproteins (i.e., apolipoproteins A, B, C and E), and/or lipoproteins (i.e., the macromolecular complexes formed by the lipid and the apolipoprotein that allow lipids to circulate in blood, such as LDL, VLDL and IDL).
  • Hyperlipidemia is associated with abnormally high levels of lipids, LDL and VLDL cholesterol, and/or triglycerides.
  • Treatment of dyslipidemia refers to the administration of the combinations of the present disclosure to a dyslipidemic subject.
  • Prevention of dyslipidemia refers to the administration of the combinations of the present disclosure to a pre-dyslipidemic subject.
  • a pre-dyslipidemic subject is a subject with higher than normal lipid levels, that is not yet dyslipid
  • LDL-C levels of 130-159 mg/dL, 160-189 mg/dL, and greater than or equal to 190 mg/dL are considered borderline high, high, and very high, respectively.
  • Total cholesterol levels of 200-239 and greater than or equal to 240 mg/dL are considered borderline high and high, respectively.
  • HDL-C levels of less than 40 mg/dL are considered low.
  • the patient has been identified as in need of lipid-lowering therapy.
  • the individual has been identified as in need of lipid-lowering therapy according to the guidelines established in 2001 by Adult Treatment Panel III (ATP III) of the National Cholesterol Education Program (NCEP), and updated in 2004 ( Grundy et al., Circulation, 2004, 110, 227-239 ).
  • ATP III Adult Treatment Panel III
  • NCEP National Cholesterol Education Program
  • the disclosure provides methods for reducing LDL-C in an individual. In certain embodiments, the disclosure provides methods for reducing VLDL-C in an individual. In certain embodiments, the disclosure provides methods for reducing IDL-C in an individual. In certain embodiments, the disclosure provides methods for reducing non-HDL-C in an individual. In certain embodiments the disclosure provides methods for reducing Lp(a) in an individual. In certain embodiments, the disclosure provides methods for reducing serum triglyceride in an individual. In certain embodiments, the disclosure provides methods for reducing liver triglyceride in an individual. In certain embodiments, the disclosure provides methods for reducing Ox-LDL-C in an individual. In certain embodiments, the disclosure provides methods for reducing small LDL particles in an individual.
  • the disclosure provides methods for reducing small VLDL particles in an individual. In certain embodiments, the disclosure provides methods for reducing phospholipids in an individual. In certain embodiments, the disclosure provides methods for reducing oxidized phospholipids in an individual.
  • An individual's response to administration of the ant-LPS immunoglobulin preparation is used by a physician to determine the amount and duration of therapeutic intervention.
  • An individual having elevated LDL-C levels may also exhibit reduced HDL-C levels and/or elevated total cholesterol levels. Individuals having elevated LDL-C levels may also exhibit elevated triglyceride levels.
  • the dyslipidemia may be characterized by a LDL cholesterol of at least 160 mg/dL, a LDL cholesterol of at least 190 mg/dL, a Total Cholesterol of at least 200 mg/dL, a Total Cholesterol of at least 240 mg/dL, a HDL Cholesterol of less than 60 mg/dL, a HDL Cholesterol of less than 40 mg/dL, serum triglycerides of between 150 and 199 mg/dL, serum triglycerides of between 200 and 499 mg/dL, or serum triglycerides of at least 500 mg/dL.
  • Treatment of dyslipidemia using the compositions of the present disclosure may result in a decrease in total cholesterol, LDL, or serum triglycerides, respectively, of about 1, 2, 3, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 and over 99% relative to untreated control, or levels prior to the treatment.
  • Measurements of cholesterol, lipoproteins and triglycerides are obtained using serum or plasma collected from an individual. Methods of obtaining serum or plasma samples are routine, as are methods of preparation of the serum samples for analysis of cholesterol, triglycerides, and other serum markers. A physician may determine the need for therapeutic intervention for individuals in cases where more or less aggressive LDL-lowering therapy is needed. The practice of the methods herein may be applied to any altered guidelines provided by the NCEP, or other entities that establish guidelines for physicians used in treating any of the diseases or conditions listed herein, for determining coronary heart disease risk and diagnosing metabolic syndrome.
  • Insulin resistance refers to a state in which a given concentration of insulin is associated with a subnormal glucose response. Insulin resistance is a component of a number of states which include, but are not limited to, the following:
  • Insulin resistance can be defined as an abnormal value according to e.g. generally known and/or accepted methods/procedures of determining insulin resistance and/or sensitivity. Those skilled in the art are well aware of certain methods/procedures of determining such insulin resistance and/or sensitivity values, including but not limited to, the homeostasis model assessment-insulin resistance (HOMA or HOMAIR), whole body insulin sensitivity index (WBISI), insulin sensitivity index (ISI), euglycemic-hyperinsulinemic clamp measure, and others.
  • HOMA or HOMAIR homeostasis model assessment-insulin resistance
  • WBISI whole body insulin sensitivity index
  • ISI insulin sensitivity index
  • euglycemic-hyperinsulinemic clamp measure euglycemic-hyperinsulinemic clamp measure
  • HOMA or HOMAIR Homeostasis model assessment-insulin resistance
  • (HOMAIR) (FI x FG)/22.5, wherein FI is the fasting insulin concentration (in microunits per milliliter) and FG is the fasting glucose lever (in millimoles per liter). Accordingly, relatively lower HOMA-IR values correspond to relatively greater insulin sensitivity, whereas relatively higher HOMA-IR values correspond to relatively lower insulin sensitivity.
  • WBISI Whole body insulin sensitivity index
  • OGTT oral glucose tolerance test
  • WBISI 10,000/square root of ((fasting glucose x fasting insulin) x (mean (OGTT) glucose x mean (OGTT) insulin)).
  • the term “insulin resistance” includes impaired glucose tolerance, impaired fasting glucose and diabetes.
  • the definition of the terms “Impaired glucose tolerance”, “impaired fasting glucose” and “diabetes” can include the clinical diagnosis definitions of the WHO, such as those published in “Definition and diagnosis of diabetes mellitus and intermediate hyperglycemia: report of a WHO/IDF consultation", (2006).
  • the term “Impaired glucose tolerance” (IGT) refers to a fasting plasma glucose of less than 7.0 mmol/l (126 mg/dl) and a 2-h plasma glucose of between 7.8 mmol/l (140 mg/dl). to less than 11.1 mmol/l (200 mg/dl).
  • a 2-hour plasma glucose refers to venous plasma glucose 2 hours after ingestion of 75g oral glucose load (Oral Glucose Tolerance Test; OGTT).
  • the term “Impaired Fasting Glucose” refers to a fasting plasma glucose of between 6.1 mmol/l (110 mg/dl) to 6.9 mmol/l (125 mg/dl), or a fasting glucose of between 6.1 mmol/l (110 mg/dl) to 6.9 mmol/l (125 mg/dl) and a 2-h plasma glucose of less than 7.8 mmol/l (140 mg/dl).
  • the term “diabetes” refers to a fasting plasma glucose of ⁇ 7.0 mmol/l (126 mg/dl) or a 2-h plasma glucose of ⁇ 11.1 mmol/l (200 mg/dl).
  • OGTT oral glucose tolerance test
  • the insulin resistance may be characterized by a fasting plasma glucose of less than 7.0 mmol/l (126 mg/dl) and a 2-h plasma glucose of between 7.8 mmol/l (140 mg/dl) to less than 11.1 mmol/l (200 mg/dl), a fasting plasma glucose of between 6.1 mmol/l (110 mg/dl) to 6.9 mmol/l (125 mg/dl), a fasting plasma glucose of between 6.1 mmol/l (110 mg/dl) to 6.9 mmol/l (125 mg/dl) and a 2-h plasma glucose of less than 7.8 mmol/l (140 mg/dl), or a fasting plasma glucose of ⁇ 7.0 mmol/l (126 mg/dl) or a 2-h plasma glucose of ⁇ 11.1 mmol/l (200 mg/dl).
  • HbA1c hemoglobin A1c
  • Improvements (e.g., decrease) in HbAlc that are indicative of therapeutic efficacy may vary depending on the initial baseline measurement in a patient, with a larger decrease often corresponding to a higher initial baseline and a smaller decrease often corresponding to a lower initial baseline.
  • the method should result in an HbAIc decrease of at least about 0.5% (e.g., at least about 0.5%, at least about 1%, at least about 1.5%, at least about 2%, at least about 2.5%, at least about 3%, at least about 3.5%, at least about 4% or more) compared with pre-dose levels.
  • An A1C of ⁇ 6.5% is indictive of diabetes.
  • GLP-I is a neuroendocrine hormone of the distal gut with a strong insulinotropic action that is synthesized and secreted from L-cells in the intestine in response to meal ingestion ( Kieffer TJ, Habener JF 1999. Endocr Rev 20: 876-913 ). Importantly, the action of GLP-I is glucose-dependent, avoiding the occurrence of hypoglycemia.
  • the intracellular precursor to GLP-I, GLP-1-(1-37) is cleaved from proglucagon, and the first six aminoacids are subsequently removed from the N terminus to form bioactive peptides.
  • GLP-1(7-36) amide about 80% of truncated GLP-I is amidated to form GLP-1(7-36) amide; the predominant secreted form of GLP-I, whereas the remainder is released as GLP-1-(7-37)( Orskov C. et al; Diabetes 43:535-539, 1994 ).
  • GLP-Ir GLP-I receptor
  • GLP- 1(7-36) NH2 and GLP- 1(7-37) have equipotent effects to stimulate glucose-stimulated insulin release, ( Orskov C. et al; Diabetes 43:535-539, 1994 ), and a physiological role for these hormones in the incretin response has been established in several animal models. Numerous effects other than stimulation of insulin release have been ascribed to GLP-I. In pancreas, it stimulates insulin biosynthesis, restoration of glucose sensitivity to the islets and stimulates increased expression of the glucose transporter GLUT-2 and glucokinase.
  • GLP-I regulates ⁇ cell mass by stimulating replication and growth and also inhibits apoptosis of existing ⁇ cells and neogenesis of new ⁇ -cells from duct precursor cells. GLP-I inhibits glucagon secretion and leads to reduced hepatic glucose output. In the gut GLP-I is a potent inhibitor of motility and gastric emptying and also inhibits gastric acid secretion. This leads to decreased food intake and reduced body weight ( Stoffers, D. A. et al; Diabetes 2000, 49, 741-748 . Drucker DJ Diabetes Care 26:2929-2940, 2003 ).
  • GLP-I acts through a G protein-coupled receptor to exert its functions. This receptor is expressed in many tissues, including pancreatic islets, the central nervous system, lung, kidney, heart, and the gut. GLP-I is coupled to its receptor through stimulatory Ga and adenylyl cyclase to increase intracellular cAMP. GLP-I can induce other intracellular signals as well, including increases in intracellular calcium, phosphoinositol 3 -kinase (PI3K) activity, and mitogen-activated protein kinase activity ( Buteau J. et al; Diabetologia 42:856-864, 1999 ; Bullock BP.et al; Endocrinology 137: 2968-2978, 1996 ).
  • PI3K phosphoinositol 3 -kinase
  • the plasma level of bioactive GLP-1 is 5-10 pmol/L in normal fasting humans, however is significantly decreased in diabetic patients.
  • Adiponectin is secreted into the bloodsteam where it accounts for approximately 0.01% of all plasma protein, at around 5-10 ⁇ g/mL. Adiponectin levels are significantly lower in the diabetic than the lean subjects, and serum adiponectin levels were statistically significantly lower in patients with NAFLD than controls.
  • treatment of insulin resistance using the compositions of the present disclosure may result in an increase in early peak of insulin secretion, adiponectin and GLP-1, of about 1, 2, 3, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 and over 99% relative to untreated control, or levels prior to the treatment.
  • Treatment of insulin resistance using the compositions of the present disclosure may result in a decrease in fasting plasma glucose, HBA1c, HOMA score, and OGTT of about 1, 2, 3, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 and over 99% relative to untreated control, or levels prior to the treatment.
  • Treatment of insulin resistance using the compositions of the present disclosure may result in an increase in early peak of insulin secretion, adiponectin and GLP-1, of about 1, 2, 3, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 and over 99% relative to untreated control, or levels prior to the treatment.
  • Nonalcoholic fatty liver disease describes a spectrum of liver diseases ranging from simple fatty liver (steatosis) to nonalcoholic steatohepatitis (NASH) with progressive fibrosis and liver failure.
  • NAFLD nonalcoholic steatohepatitis
  • hyperglycemia with or without evidence of hyperlipidemia is commonly associated with NAFLD.
  • the disease exhibits the histological features of alcohol-induced liver disease in patients who do not consume significant amounts of alcohol. All of the stages of NAFLD have in common the accumulation of fat in the liver cells.
  • Farrell and Larter in Hepatology, 243:899S112 (2006 ) describe NASH as "the lynchpin" between hepatic steatosis and cirrhosis in the spectrum of NAFLD.
  • Alcoholic hepatitis is a precursor to cirrhosis and is caused by alcohol.
  • the typical histologic picture includes hepatocellular necrosis and ballooning degeneration, and alcoholic Mallory's hyaline bodies (abnormal aggregations of cellular intermediate filament proteins indicative of fibrosis). Cholestasis is prominent.
  • Alcoholic hepatitis can range from a mild hepatitis, with abnormal laboratory tests being the only indication of disease, to severe liver dysfunction with complications such as jaundice (yellow skin caused by bilirubin retention), hepatic encephalopathy (neurological dysfunction caused by liver failure), ascites (fluid accumulation in the abdomen), bleeding esophageal varices (varicose veins in the esophagus), abnormal blood clotting and coma. Alcoholic hepatitis is reversible if the patient stops drinking, but it usually takes several months to resolve. Alcoholic hepatitis can lead to liver scarring and cirrhosis.
  • the present disclosure provides methods of treatment and/or prophylaxis of alcoholic hepatitis.
  • ALT alanine aminotransferase
  • gamma-GT gamma-glutamyltransferase
  • a human subject in need of treatment may also be presumptively diagnosed by serum tests of liver enzymes.
  • steatosis may be indicated by elevated serum levels (often moderately elevated, e.g., elevated approximately 2, 3, 4, 5, 6, 7, 9, 10, 11, or 12-fold above normal levels) of liver enzymes (such as, e.g., alanine aminotransferase, aspartate aminotransferase, ⁇ -glutamyltransferase, alkaline phosphatase) when other causes (such as, e.g., acute hepatitis, autoimmune disease, chronic hepatitis, cirrhosis, fulminant hepatitis, hepatocellular carcinoma, metastatic carcinoma, right heart failure, and viral hepatitis) have been eliminated.
  • liver enzymes such as, e.g., alanine aminotransferase, aspartate aminotransferase, ⁇ -glutamyltransferase, alkaline
  • alanine aminotransferase (ALT or SGPT) values greater than 32, 24, or 56 units per liter of serum or at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more times normal values may be indicative of a disorder associated with hepatic lipid deposits, or by aspartate aminotransferase (AST or SGOT) values greater than 40 units per liter of serum or at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more times normal values.
  • AST or SGOT aspartate aminotransferase
  • the ratio of AST to ALT is often less than one in NAFLD, but may be greater than one in patients with alcoholic liver disease or advanced liver disease.
  • ⁇ -glutamyltransferase levels may be significantly elevated, e.g., at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more times normal values.
  • the mean corpuscular volume (MPV) may be greater than, e.g., 86, 98, 100, or 110 femtoliters.
  • elevated liver enzymes relates to elevated levels of liver enzymes such as Alanine transaminase (ALT) g-glutamyl-transferase (GGT), Aspartate transaminase (AST), and Alkaline phosphatase (ALP).
  • ALT Alanine transaminase
  • GTT g-glutamyl-transferase
  • AST Aspartate transaminase
  • ALP Alkaline phosphatase
  • the degree of enzyme elevation may be between 1 and 4 times the upper limit of normal values.
  • the elevated liver enzymes may be characterized by an AST of greater than 40 IU/L, ALT of greater than 30 IU/L, and ALT of greater than 56 IU/L, an ALP of greater than 115 IU/L, or a GGT of greater than 80 IU/L.
  • a subject in need of treatment may also be presumptively diagnosed by noninvasive imaging techniques (e.g., ultrasonography, computed tomography, and magnetic resonance imaging) when steatosis is greater than, e.g., 25% or 30%.
  • noninvasive imaging techniques e.g., ultrasonography, computed tomography, and magnetic resonance imaging
  • NAFLD may present as a focal or diffuse accumulation of lipid, but in NASH the lipid is generally diffuse.
  • NAFLD may also be detected by magnetic resonance spectroscopy, a technique which may be of value for quantitative determination of hepatic lipid levels.
  • a subject in need of treatment may be definitively diagnosed by liver biopsy.
  • a liver is considered to be steatotic when a biopsy reveals at least 5-10% w/w fatty deposits (in practice, this is value may be determined microscopically as the fraction of lipid-filled hepatocytes). See, e.g., Clark et al., J. Am. Med. Assoc. 289:3000-3004 (2003 ) and Adams et al., Can. Med. Assoc. J. 172:899-905 (2005 ).
  • a liver with fatty deposits comprising up to 25% w/w may be considered mildly steatotic, and a liver with fatty deposits comprising greater than 25% w/w may be considered severely steatotic.
  • Histological findings indicative of NASH include steatosis, hepatocyte ballooning, lobular inflammation, Mallory hyaline bodies, mixed inflammatory infiltrate, pericellular fibrosis, and perisinusoidal fibrosis. Additional information may be found in, e.g., Neuschwander-Tetri et al., Hepatology 37:1202-1219 (2003 ).
  • NAFLD/NASH Disease progression in NAFLD/NASH, as assessed by fibrosis in liver histology, has been reported to correlate with the degree of insulin resistance and other features of metabolic syndrome.
  • Other markers proposed to be related to fibrosis in NAFLD patients include laminin, hyaluronan, type IV collagen, and aspartate aminotransferase.
  • Female gender is also associated with more rapid disease progression.
  • the fatty liver may be characterized by macrovesicular steatosis, macrovesicular steatosis and necroinflammatory activity, or a NAS score of at least 4.
  • Efficacy of treatment may also be determined by detection of a reduction in one or more symptoms or clinical manifestations of a disease as well as any of the tests described above for diagnosis.
  • Treatment of liver damage/fatty liver using the compositions of the present disclosure may result in a decrease AST, ALT, AP, GGT about 1, 2, 3, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 and over 99% relative to untreated control, or levels prior to the treatment.
  • One or more of the following secondary endpoints also may be determined in order to assess efficacy of the treatment, such as for example fasting blood sugar (e.g., glucose) levels (e.g., decrease to ⁇ 130, ⁇ 125, ⁇ 120, ⁇ 115, ⁇ 110, ⁇ 105, ⁇ 100; alternatively decrease of >20%, >30%, >40%, >50%, >60%, >70%, >80%, >90%, >95% compared to pre-dose levels), 120 minute oral glucose tolerance test (OGTT) (e.g., ⁇ 200, ⁇ 190, ⁇ 180, ⁇ 170, ⁇ 160, ⁇ 150, ⁇ 140), glucose/insulin C-peptide AUC (e.g., >25%, >50%, >60%, >70%, >80%, >90%, >100% increase from pre-treatment), reduction in diabetes medication (e.g., insulin, oral hypoglycemic agent), improvement in insulin sensitivity, serum cytokine levels (e.g.
  • the effect on hyperglycemia can be assessed by measuring fasting blood sugar (i.e., glucose) levels
  • the effect on hyperinsulinemia may be assessed by measuring insulin levels and/or C-peptide levels
  • the effect on obesity may be assessed by measuring weight and/or BMI
  • the effect on insulin resistance may be assessed by OGTT.
  • subjects treated in accordance with the present disclosure may experience a decrease in a cardiovascular risk indicator(s) and/or a decrease in serum lipids with improvement in the lipid profile.
  • Such measurements of serum lipids and/or lipid profile may include, for example a decrease in cholesterol, a decrease in low-density lipoprotein cholesterol (LDL), a decrease in very-low-density lipoprotein cholesterol (VLDL), a decrease in triglycerides, a decrease in free fatty acids, a decrease in apolipoprotein B (Apo B), an increase in high-density lipoprotein cholesterol (HDL), maintaining the level of high-density lipoprotein cholesterol (HDL) compared to pre-treatment level, and/or an increase in apolipoprotein A (Apo A).
  • LDL low-density lipoprotein cholesterol
  • VLDL very-low-density lipoprotein cholesterol
  • HDL high-density lipoprotein cholesterol
  • HDL high-density lip
  • a decrease in the level of cholesterol may be a decrease of at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, or more from the pre-treatment level.
  • a decrease in the level of low-density lipoprotein cholesterol may be a decrease of at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, or more from the pre-treatment level.
  • a decrease in the triglyceride level in the blood of the subject may be a decrease of at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or more from the pre-treatment level.
  • a decrease in the level of free fatty acids may be a decrease of at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, or more from the pre-treatment level.
  • An increase in the level of high-density lipoprotein cholesterol may be an increase of at least 1%, 2%, 3%, 4%, 5%, 6%, 8%, 10%, 12%, 14%, 16%, or more from the pre-treatment level.
  • subjects treated in accordance with the present disclosure may experience a decrease in insulin resistance.
  • Such decrease in insulin resistance may be measured by an improvement in a homeostasis model assessment (HOMA), an insulin tolerance test, an insulin suppression test, a steady-state plasma glucose method, or any of the other assay methods know in the art (see for example Matthews et al, 1985, Diabetologia 28:412-419 ; Odegaard et al., 2007, Nature 447:1116-1121 ; Emoto et al., 1999, Diabetes Care 22:818-822 ).
  • Other of the aforementioned measurements may be made using any of a variety of standard assays known in the art, for example assays published in Chernecky CC, Berger BJ, eds.
  • the anti-LPS immunoglobulin preparation may be derived from colostrum or avian eggs.
  • composition of the present disclosure in order to practice the present methods of therapy is carried out by administering a therapeutically effective amount of the compounds in the composition to a subject in need of such treatment or prophylaxis.
  • the need for a prophylactic administration according to the methods of the present disclosure is determined via the use of the well known risk factors as described herein.
  • Each oral dose form may, for example, comprise the colostrum equivalent of less than 1200 mg (dry weight basis), preferably 800 mg, preferably less than 400 mg, more preferably less than 200 mg.
  • colostrum equivalent we mean the amount of raw colostrum, howsoever purified, which is processed to provide the contents of a dose form.
  • the oral dose form may comprise 5 mg to 500 mg bovine colostrum powder (BCP) (dry weight basis), e.g. 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 125, 150, 175, 200, 225, 250, or 500 mg.
  • BCP bovine colostrum powder
  • the oral dose form may comprise 500 mg to 5000 mg bovine colostrum powder (dry weight basis), e.g. 500, 750, 1000, 1250, 1500, 1750, 2000, 2250, 2500, 2750, 3000, 3250, 3500, 3750, 4000, 4250, 4500, 4750 or 5000 mg.
  • bovine colostrum powder dry weight basis
  • Suitable dosage ranges are, e.g. from about 5mg to about 5000mg/day, preferably 50mg to about 5000mg/day, more preferably 500mg to about 5000mg/day, or most preferably 1500mg to about 2000mg/day BCP (dry weight basis). In one preferred embodiment, the dose is 1800mg/day BCP (dry weight basis).
  • the antibodies are present in the composition for oral administration in an amount sufficient to provide from at least 7% by dry weight of the composition of IgG.
  • the antibodies are present in the composition for oral administration in an amount sufficient to provide from at least 40% by weight of the composition of IgG.
  • the oral dose form may comprise 2 mg to 200 mg IgG, e.g. 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 125, 150, 175, or 200 mg IgG.
  • the oral dose form may comprise 200 mg to 2000 mg IgG e.g. 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900 or 2000 mg IgG.
  • antibodies specific to the antigen are present in the composition for oral administration in an amount sufficient to provide from at least 10% specific IgG of the weight of IgG.
  • the oral dose form may comprise .2 mg to 20 mg specific IgG, e.g..2, .4, .6, .8, 1, 1.2, 1.4, 1.6, 1.8, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10.0, 11.0, 12.5, 15.0, 17.5, or 20.0 mg specific IgG.
  • specific IgG e.g..2, .4, .6, .8, 1, 1.2, 1.4, 1.6, 1.8, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10.0, 11.0, 12.5, 15.0, 17.5, or 20.0 mg specific IgG.
  • Suitable dosage ranges are, e.g. from about 2 to about 200 mg/day, preferably 20 to about 2000 mg/day, more preferably 200 to about 2000mg/day, or most preferably 600 mg to about 800 mg/day IgG. In one preferred embodiment, the dose is 720mg/day IgG.
  • the anti-LPS immunoglobulin preparation is not administered at a dose of about 600mg per day (dry weight volume).
  • the oral dose form may be administered for about 1, 2, 3, 4, 5, 6,7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or 31 days.
  • the oral dose form is administered for 30 days.
  • the anti-LPS immunoglobulin preparation is administered for 30 days at dose of 1.8g/day. In one embodiment, the anti-insulin immunoglobulin preparation is administered for 30 days at dose of 1.2g/day.
  • the oral dose form preferably comprises colostrum derived from the hyperimmune colostrum and/or colostrum which has been added to the polyclonal antibodies in accordance with the teaching of PCT/AU03/00348 (Pub. No.: WO/2003/080082 ).
  • the oral dosage form may also comprise a buffer system such as that disclosed in PCT/AU2005/001746 (Pub. No.: WO/2006/053383 ).
  • terapéuticaally effective amount means the amount of the active compounds in the composition that will elicit the biological or medical response in a tissue, system, subject, or human that is being sought by the researcher, veterinarian, medical doctor or other clinician, which includes alleviation of the symptoms of the disorder being treated.
  • the novel methods of treatment of this disclosure are for disorders known to those skilled in the art.
  • prophylactically effective amount means the amount of the active compounds in the composition that will elicit the biological or medical response in a tissue, system, subject, or human that is being sought by the researcher, veterinarian, medical doctor or other clinician, to prevent the onset of the symptoms of the disorder in a subject at risk of developing the disorder.
  • prophylactic or therapeutic dose of the active ingredients can, of course, vary with the nature of the severity of the condition to be treated. It can also vary according to the age, weight and response of the individual patient, and may be administered in subject in single or divided doses. On the other hand, it may be necessary to use dosages outside the ranges provided herein in some cases.
  • the anti-LPS immunoglobulin preparation may be administered at a dose of about 5 mg to about 25000 mg per day, 10 mg to about 20000 mg per day, 25 mg to about 15000 mg per day, 100 mg to about 2000 mg per day, or about 1800 mg per day. In one embodiment, the anti-LPS immunoglobulin preparation is not administered at a dose of about 600 mg per day.
  • the anti-LPS immunoglobulin preparation may be formulated for administration at a dose of about 5 mg to about 25000 mg per day, about 10 mg to about 20000 mg per day, about 25 mg to about 15000 mg per day, about 100 mg to about 2000 mg per day or about 1800 mg per day.
  • the anti-LPS immunoglobulin preparation is not formulated for administration at a dose of about 600 mg per day.
  • the bacteria from which each type of O antigen is isolated are grown in separate bacterial culture systems, and after separation of the O antigen from the bacteria, the component antigens are added together to form a component of the vaccine.
  • Methods of preparing LPS/O antigen are known in the art and described in WO/2004/078209 .
  • Methods of preparing hyperimmune bovine colostrum (HIBC) are also described in WO/2004/078209 .
  • the anti-LPS immunoglobulin preparation may be prepared by immunizing a mammal or avian with LPS from multiple E. coli strains.
  • the mammal or avian may be immunized with LPS selected from the group consisting of 06, 08, 015, 025, 027, 063, 078, 0114, 0115, 0128, 0148, 0153, 0159, and other LPS associated with enterotoxigenic E. coli.
  • the mammal or avian may be immunized with LPS selected from the group consisting of O78, O6, O8, O129 and 0153 LPS.
  • LPS may comprise 078 LPS.
  • composition further comprises an anti-insulin immunoglobulin preparation.
  • the anti-insulin immunoglobulin preparation is administered at a dose of about 5mg to about 25000mg per day, about 10 mg to about 20000 mg per day, about 25 mg to about 15000 mg per day, about 50 mg to about 10000 mg per day, about 50 mg to about 4000mg per day, about 500 mg to about 3000mg per day, about 1000 mg to about 1400mg per day, or about 1200 mg per day.
  • the anti-insulin immunoglobulin preparation may be formulated for administration at a dose of about 5mg to about 25000mg per day, about 10 mg to about 20000 mg per day, about 25 mg to about 15000 mg per day, about 50 mg to about 10000 mg per day, about 50 mg to about 4000mg per day, about 500 mg to about 3000mg per day, about 1000 mg to about 1400mg per day or of about 1200 mg per day.
  • the oral dose form may comprise 5 mg to 500 mg bovine colostrum powder (BCP) (dry weight basis), e.g. 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 125, 150, 175, 200, 225, 250, or 500 mg.
  • BCP bovine colostrum powder
  • the oral dose form may comprise 500 mg to 5000 mg bovine colostrum powder (dry weight basis), e.g. 500, 750, 1000, 1250, 1500, 1750, 2000, 2250, 2500, 2750, 3000, 3250, 3500, 3750, 4000, 4250, 4500, 4750 or 5000 mg.
  • bovine colostrum powder dry weight basis
  • the antibodies are present in the composition for oral administration in an amount sufficient to provide from at least 7% by dry weight of the composition of IgG.
  • the antibodies are present in the composition for oral administration in an amount sufficient to provide from at least 40% by weight of the composition of IgG.
  • the oral dose form may comprise 2 mg to 200 mg IgG, e.g. 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 125, 150, 175, or 200 mg IgG.
  • the oral dose form may comprise 200 mg to 2000 mg IgG e.g. 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900 or 2000 mg IgG.
  • antibodies specific to the antigen are present in the composition for oral administration in an amount sufficient to provide from at least 10% specific IgG of the weight of IgG.
  • the oral dose form may comprise .2 mg to 20 mg specific IgG, e.g..2, .4, .6, .8, 1, 1.2, 1.4, 1.6, 1.8, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10.0, 11.0, 12.5, 15.0, 17.5, or 20.0 mg specific IgG.
  • specific IgG e.g..2, .4, .6, .8, 1, 1.2, 1.4, 1.6, 1.8, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10.0, 11.0, 12.5, 15.0, 17.5, or 20.0 mg specific IgG.
  • the oral dose form may be administered for about 1, 2, 3, 4, 5, 6,7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or 31 days.
  • the anti-insulin immunoglobulin preparation may be prepared by immunizing a mammal or avian with insulin conjugated to a protein.
  • KLH keyhole limpet hemocyanin
  • serum albumin serum albumin
  • bovine thyroglobulin bovine thyroglobulin
  • adjuvants examples include Freund's complete adjuvant and MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate).
  • the immunization protocol may be selected by one skilled in the art without undue experimentation.
  • the anti-insulin immunoglobulin preparation may be prepared by immunizing a mammal or avian with insulin conjugated to keyhole limpet hemocyanin (KLH).
  • KLH keyhole limpet hemocyanin
  • the present disclosure provides a method for reducing fasting glucose levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for increasing the early peak of insulin secretion in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing oral glucose tolerance in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for increasing insulin secretion in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing HBA1C levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing triglyceride levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing total cholesterol levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing LDL cholesterol levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing ALT levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing AST levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing ALP levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing GGT levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for increasing GLP-1 levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for increasing Adiponectin levels in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for increasing the Adiponectin/IL-6 ratio in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for increasing the CD25+ T regulatory cells in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing body weight in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a method for decreasing waist circumference or arm circumference in a human patient in need thereof, comprising administering to said patient a therapeutically effective amount a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in reducing fasting glucose levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in increasing the early peak of insulin secretion in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing oral glucose tolerance in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in increasing insulin secretion in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing HBA1C levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing triglyceride levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing total cholesterol levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing LDL cholesterol levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing ALT levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing AST levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing ALP levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing GGT levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in increasing GLP-1 levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in increasing Adiponectin levels in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in increasing the Adiponectin/IL-6 ratio in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in increasing the CD25+ T regulatory cells in a human patient in need thereof
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing body weight in a human patient in need thereof.
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in decreasing waist circumference or arm circumference in a human patient in need thereof.
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for reducing fasting glucose levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for increasing the early peak of insulin secretion in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing oral glucose tolerance in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for increasing insulin secretion in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing HBA1C levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing triglyceride levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing total cholesterol levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing LDL cholesterol levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing ALT levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing AST levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing ALP levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing GGT levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for increasing GLP-1 levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for increasing Adiponectin levels in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for increasing the Adiponectin/IL-6 ratio in a human patient in need thereof
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for increasing the CD25+ T regulatory cells in a human patient in need thereof
  • Treatment using the compositions of the present disclosure may result in an increase in T regulatory cells, of about 1, 2, 3, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 and over 99% relative to untreated control, or levels prior to the treatment.
  • T cell responses may be quantified using methods known in the art, for example ELISPOT assays, flow cytometry or useful immunodetection methods described in the scientific literature, such as, e.g., Maggio et al., Enzyme-Immunoassay, (1987 ) and Nakamura, et al., Enzyme Immunoassays: Heterogeneous and Homogeneous Systems, Handbook of Experimental Immunology, Vol. 1: Immunochemistry, 27.1-27.20 (1986 . Immunoassays, in their most simple and direct sense, are binding assays involving binding between antibodies and antigen. Many types and formats of immunoassays are known and all are suitable for detecting the disclosed T regulatory cells.
  • immunoassays are enzyme linked immunosorbent assays (ELISAs), enzyme linked immunospot assay (ELISPOT), radioimmunoassays (RIA), radioimmune precipitation assays (RIPA), immunobead capture assays, Western blotting, dot blotting, gel-shift assays, Flow cytometry, protein arrays, multiplexed bead arrays, magnetic capture, in vivo imaging, fluorescence resonance energy transfer (FRET), and fluorescence recovery/localization after photobleaching (FRAP/ FLAP)
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing body weight in a human patient in need thereof.
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for decreasing waist circumference or arm circumference in a human patient in need thereof.
  • the present disclosure provides a method of treating a human suffering a T-cell mediated disease comprising administering to the human an effective amount of a composition comprising an anti-LPS immunoglobulin preparation.
  • T cell mediated disease is intended to mean a condition in which an inappropriate T cell response is a component of the pathology.
  • the term is intended to include both diseases directly mediated by T cells and those indirectly mediated by T cells such as diseases in which an inappropriate T cell response contributes to damage resulting from the production of autoimmune antibodies.
  • the term is intended to encompass both T cell mediated autoimmune diseases and unregulated clonal T cell replication. Therefore, a T cell mediated disease includes T cell mediated conditions exhibiting clinically recognizable symptoms as well as T cell mediated dysfunctions.
  • T cell mediated diseases include type 1 diabetes, insulitis, Graves' disease, rheumatoid arthritis (RA), multiple sclerosis (MS), systemic lupus erythematosus, myasthenia gravis, pemphigus vulgaris, Hashimoto's Thyroditis, Autoimmune Uveitis, Sjogren's syndrome, Dermamyositis and Addison's disease.
  • Thl-mediated in reference to a disease, disorder, or condition is one that has been associated with increased cytokine production from Th1 cells, including IFN-Y, IL-2, GM-CSF, TNF-alpha and IL-3. Specific examples include multiple sclerosis, insulindependent diabetes mellitus, Crohn's disease, uveitis, chronic rheumatism, and systemic lupus erythematosus.
  • Th2 mediated in reference to a disease, disorder, or condition is one that has not been associated with increased cytokine production from Th1 cells.
  • Specific examples include scleroderma, multiple myositis, vasculitis syndrome, mixed connective tissue disease, Sjogren's syndrome, hyperthyroidism, Hashimoto's disease, myasthenia gravis, Guillain-Barre syndrome, autoimmune hepatopathy, ulcerative colitis, autoimmune nephropathy, autoimmune hematopathy, idiopathic interstitial pneumonia, hypersensitivity pneumonitis, autoimmune dermatosis, autoimmune cardiopathy, autoimmune infertility, and Behcet's disease.
  • autoimmune disease(s) is used herein to refer to a large group of illnesses, some with ill-defined causes, thought to be associated with abnormalities in immunoregulation. Therefore, the term as used herein is intended to include, but is not limited to, diseases such as rheumatoid arthritis, lupus, graft versus host disease, host versus graft disease, insulindependent diabetes, autoimmune encephlomyelitis, autoimmune hepatitis, Crohn's disease, and multiple sclerosis.
  • allergy is meant to encompass allergic disease(s) including, but not limited to, chronic bronchitis, atopic dermatitis, pollinosis (allergic rhinitis), allergic angiitis, allergic conjunctivitis, allergic gastroenteritis, allergic hepatopathy, allergic cystitis, and allergic purpura.
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in treating a human suffering a T-cell mediated disease
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament for the treatment and/or prophylaxis of a human subject suffering a T-cell mediated disease
  • the T-cell mediated disease may be insulin resistance, impaired glucose tolerance, diabetes, metabolic syndrome, or a disease associated therewith, or non-alcoholic steatohepatitis (NASH).
  • NASH non-alcoholic steatohepatitis
  • the present disclosure provides a method of treatment of a human suffering a disease selected from insulin resistance or associated disorders comprising administering an effective amount of a composition comprising an anti-LPS immunoglobulin preparation.
  • the present disclosure provides a composition comprising an anti-LPS immunoglobulin preparation for use in treatment of a human suffering a disease selected from insulin resistance or associated disorders.
  • the present disclosure provides a use of an anti-LPS immunoglobulin preparation in the manufacture of a medicament treatment of a human suffering a disease selected from insulin resistance or associated disorders.
  • the insulin resistance or an associated disorder may be diabetes, metabolic syndrome or non-alcoholic steatohepatitis (NASH).
  • diabetes metabolic syndrome or non-alcoholic steatohepatitis (NASH).
  • NASH non-alcoholic steatohepatitis
  • the product 'BioGARD' is a colostrum preparation supplied by Immuron Limited.
  • Each Immuron BioGARD tablet is an uncoated 1.2g oral tablet, which contains 600 mg of freeze-dried Bovine Colostrum Powder, in combination with excipients.
  • the active substance in BioGARD tablets is freeze-dried bovine colostrum powder milked from commercial dairy cowherds. The cows in these herds, as well as being vaccinated for routine cattle pathogens, have been vaccinated with a vaccine against the outer cell wall antigens of multiple strains of E. coli bacteria, a major organism in human gut microflora.
  • Immuron BCP (bovine colostrum powder) is a high-protein (>80%), lactose- and fat-reduced natural product derived from the first milking of commercial dairy cows collected after calving. It is presented before tableting as a concentrated, freeze dried powder.
  • Immuron BCP contains approximately 40% antibodies (immunoglobulins) in the dry powder.
  • the immunoglobulins in BioGARD's BCP have high binding activity against the Lipopolysaccharide (LPS) of Gram-negative bacteria. Binding of LPS is assayed by Immuron using a standardized ELISA and immuno-blotting detection systems.
  • BioGARD was produced as follows. Dairy cows were immunized with a mixture of LPS antigens. For example, dairy cows were immunized with LPS 078, or a mixture of 06, 08, 015, 025, 027, 063, 078, 0114, 0115, 0128, 0148, 0153, 0159, and other LPS associated with enterotoxigenic E. coli.
  • the antigen vaccine was administered during the last eight weeks of gestation.
  • Colostral milk was collected during the first two days of lactation. The milk fat was removed and skim milk was pasteurized at 56° C for 30 minutes and then coagulated by renetting as in Hilpert, Human Milk Banking 1984.
  • the precipitate was dissolved in 0.01M TRIS-HCl buffer pH 8 containing 0.32M NaCl (30% of original volume). This solution was extensively dialyzed against five volumes of the same buffer using an Amicon spiral membrane SIY30 cartridge. The antibody solution was then concentrated to 10%, snap frozen and freeze dried.
  • Antibody fragments are prepared according to modified method based on the methods described by Jones R.G.A. and Landon J. ( Jones R.G.A. and Landon J. J. Immunol. Methods 263: 57-74 (2002 )). Briefly, an equal volume of 0.2M Sodium Acetate buffer pH 4.0, is added to a colostrum pool obtained from immunized animals as described above. The pH of the diluted colostrum pool has been adjusted to 4.6 and incubated at 37°C for two hours to precipitate caseins. Subsequently, colostrum is centrifuged and filtered (0.45 ⁇ m) to remove casein.
  • the pH of the resultant colostral whey has been adjusted to pH 4.0, followed by addition of Pepsin (Enzyme Solutions with 1:15,000 activity) at 5.0% w/w and incubation for twenty hours at 45°C. Pepsin digestion has been stopped by addition of 0.5 vol. of IM Tris pH8 and cooling the reaction mix to 4°C. The pH of the reaction is adjusted to pH to 7.0 and the F(ab')2 mix is concentrated using 30kD ultra-filtration membrane and dia-filtrate vs. >50 volumes of 20mM sodium phosphate/150mM NaCl pH 6.0 buffer.
  • Fab ' by 2-mercaptoethylamine (MEA).
  • MEA 2-mercaptoethylamine
  • the mixture is then incubated at 37°C for 90mins.
  • the reaction mixture is applied on a PD-10 column, or a suitable G25 column, to remove the excess MEA.
  • 0.1M sodium phosphate (pH 6.0, with 5mM EDTA-disodium) is used as the running buffer.
  • the first protein peak which contains Fab' is collected and used for treating the corresponding different indications as indicated herein below.
  • colostrum was collected from approximately 200 commercial dairy cowherds, The cows in these herds, as well as being vaccinated for routine cattle pathogens, have been vaccinated with a vaccine against the outer cell wall antigens of multiple strains of E. coli bacteria, a major organism in human gut microflora. The obtained colostrum was frozen in individual bags for testing. For processing, colostrum was thawed, pooled and fat was removed. Each batch was pasteurized. Colostrum was concentrated by ultra-filtration to reduce volume before freeze drying. The ultra-filtration step reduced lactose in the final powder to less than 7% (from about 50%).
  • Example 2 Use of colostrum-derived anti-LPS enriched immunoglobulin preparations in the treatment of hepatitis
  • mice For immune mediated hepatitis model, eleven to twelve weeks old male C57/bl mice are tail vein injected with a dose of 500 ⁇ g/mouse (approximately 15 mg/kg) of Con A (MP Biomedicals, USA) which is dissolved in 50mM Trig pH 7, 150mM NaCl, 4mM CaCl 2 , known to induce hepatitis. Animals of all tested groups are orally administered using different concentrations and preparations of specific antibodies, or the BioGARD preparation described in experimental procedures, as compared to untreated controls.
  • Con A MP Biomedicals, USA
  • AST serum aspartate aminotransferase
  • ALT alanine aminotransferase
  • histological examination of liver specimens FACS analysis of intrahepatic and intrasplenic lymphocytes for NKT markers, measurement of serum cytokine levels and Western blot analysis for the expression of the transcription factors STAT 1, 4 and 6 and NF ⁇ B and are compared to control groups.
  • Example 3 Oral administration of colostrum enriched with anti LPS antibodies
  • colostrum was collected from approximately 200 commercial dairy cowherds, The cows in these herds, as well as being vaccinated for routine cattle pathogens, have been vaccinated with a vaccine against the outer cell wall antigens of multiple strains of E. coli bacteria, a major organism in human gut microflora.
  • the obtained colostrum was frozen in individual bags for testing.
  • colostrum was thawed, pooled and fat was removed. Each batch was pasteurized. Colostrum was concentrated by ultra-filtration to reduce volume before freeze drying. The ultra-filtration step reduced lactose in the final powder to less than 7% (from about 50%).
  • compositions comprising anti-LPS immunoglobulin preparations (hyperimmune bovine colostrum) were prepared by vaccinating pregnant cows with bacterial cell wall antigens (e.g. LPS) prepared from a number of E. coli strains to produce highly concentrated antibodies (including IgG) to LPS in colostrum.
  • bacterial cell wall antigens e.g. LPS
  • this anti-LPS immunoglobulin preparation is denoted by 'HIBC'.
  • a second preparation was prepared by vaccinating pregnant cows with a vaccine comprising a number of E. coli strains, and also enriched for LPS and other surface antigens, to produce highly concentrated antibodies (including IgG) to LPS in colostrum. IgG was then purified from this colostrum preparation.
  • this composition is denoted by 'T-IgG'
  • mice Two groups of mice (Table 1) were studied. Mice (10 per group) were fed (perorally) daily for 7 days with 30ul of water (control, group A) or 30ul (approximately 3mg) of anti-LPS enriched colostrum-derived immunoglobulin preparation (group B) which was dissolved in water. After 7 days mice were sacrificed. On sacrifice day, cardiac blood was collected by standard techniques then serum was obtained for future purposes.
  • mice Naive C57Bl/6 mice (age 11-12 weeks) were used. Mice were obtained from Harlan Laboratories (Jerusalem, Israel) and were maintained in the Animal Core of the Hadassah-Hebrew University Medical School. Mice were administered standard laboratory chow and water ad libitum and kept in a 12-hour light/dark cycle. The animal experiments were carried out according to the guidelines of the Hebrew University-Hadassah Institutional Committee for Care and Use of Laboratory Animals and with the committee's approval.
  • livers After harvesting the livers are transferred to ice cold PBS cut, minced and homogenized using a dounce homogenizer with 9ml of sterile cold lysis buffer 1, divided into eppendorff tubes (1.5ml in each tube), and kept on ice for 30 minutes, followed by sonication (five cycles of 25 seconds) and centrifugation (at 4°C, 14,000 RPM for 15 minutes). The supernatants are collected into one tube, sampled for protein quantification using the Bradford technique and stored at -20°C.
  • Spleen Following excision the spleens are minced on cells dissociation grids (60 mesh) in RPMI 1640 medium, centrifuged (at 4°C, 1,400 RPM for 10 minutes) and the supernatant discarded; Red blood cells are lysed by adding 1ml of cold RBC lysis buffer (155mM ammonium chloride), followed by rinsing three times with cold PBS and centrifugation.
  • cold RBC lysis buffer 155mM ammonium chloride
  • Isolation of splenic and hepatic lymphocytes for determination of T cell subpopulations Mice of different experimental models are sacrificed on the days indicated. Splenic lymphocytes and NKT cells are isolated and red blood cells removed. Intrahepatic lymphocytes are isolated as follows: After cutting the inferior vena cava above the diaphragm, the liver is flushed with cold PBS until it become pale, followed by removal of connective tissue and gall bladder. Livers and spleens were kept in RPMI-1640 + FCS. Then spleens were crushed through a 70 ⁇ m nylon cell strainer (Falcon) and centrifuged (1250 rpm for 7 min) for the removal of cell debris.
  • Falcon 70 ⁇ m nylon cell strainer
  • Red blood cells were lysed with 1ml of cold 155mM ammonium chloride lysis buffer and immediately centrifuged (1250 rpm for 3min). Splenocytes were then washed and resuspended with ImI RPMI + FCS. Remains of connective tissue were removed. The viability by trypan blue staining was above 90%.
  • livers were first crushed through a stainless mesh (size 60, Sigma) and the cell suspension was placed in a 50-ml tube for 5min to enable cell debris to descend. 10ml of Lymphoprep (Ficoll, Axis-Shield PoC AS, Oslo, Norway) was slowly placed under the same volume of cell suspension in 50-ml tubes.
  • the tubes were then centrifuged at 1800 rpm for 18min. Cells in the interface were collected and moved to new tubes which were centrifuged again at 1800 rpm for 10 min, to obtain a pellet of cells depleted of hepatocytes to a final volume of 250 ⁇ l. Approximately 1x10 6 cells/mouse liver, were recovered. Cells viability was detected by trypan blue staining.
  • Adipose tissue (visceral fat pads) was minced and incubated in Krebs- Ringer bicarbonate buffer (3 mL/g adipose tissue) containing 10 mM glucose and 2.5% bovine serum albumin, incubated with 840 U/g collagenase type I (Sigma, Rehovot, Israel) at 37 °C with gentle agitation for 1 hour. Then filtered twice through chiffon mesh (100 ⁇ m) and centrifuged 50xg for 5 minutes. Floating adipocytes were then separated from the pellet of stromal vasculature (SV) fraction. The lower fraction was removed and centrifuged at 200xg for 5 min to pellet the SV cells. Cell number was then counted.
  • SV stromal vasculature
  • Flow cytometery analysis for determination of different subsets of lymphocytes. Following lymphocyte isolation from blood, spleen or any organ, triplicates of 2-5X10 5 cells/500 ⁇ L PBS are placed in Falcon 2052 tubes, incubated with 4 mL of 1% BSA for 10 minutes, and centrifuged at 1400 rpm for 5 minutes. Cells are re-suspended in 10 ⁇ L FCS with 1:20 labeled (FITC, APC or PE-labeled) primary antibodies directed to the following lymphocyte markers: CD3, CD4, CD8, NK1.1, CD25, FOX p3, LAP cells, IL- 17, Annexiin and surface markers for T cell activation.
  • FITC labeled
  • Cells-antibody mixtures are mixed every 10 minutes for 30 minutes.
  • Cells are isolated using anti-CD3 and anti-CD4, anti-CD8, and anti-NK1.1, respectively.
  • Cells are washed twice in 1% BSA and kept at 4°C until reading. For the control group, only 5 ⁇ L of 1% BSA are added.
  • Surface staining was performed by incubating cells with antibodies and anti-CD 16/32 (blocks Fc, eBioscience) at 4°C in FACS buffer containing PBS and 0.5% BSA, for 30 min. Cells were further washed twice with FACS buffer, resuspended in FACS buffer, and analyzed by flow cytometry.
  • Analytical cell sorting is performed on IxI0 4 cells from each group with a fluorescence-activated cell sorter (FACStar Plus, Becton Dickinson). Appropriate isotype controls were used in all experiments. Analysis was performed using a FACSCalibur instrument (Becton Dickinson, San Jose, CA). Only live cells were counted, and background fluorescence from non-antibody-treated lymphocytes was subtracted. Gates were set on forward- and side-scatters to exclude dead cells and red blood cells. Data was analyzed by the Consort 30 two-color contour plot (Becton Dickinson, Oxnard, CA, USA) or Cell Quest programs.
  • NKT lymphocyte percentage FACS analysis for determination of NKT lymphocyte percentage. Immediately after lymphocyte isolation, triplicates of 2-5x10 4 cells/500 ⁇ l PBS are placed into Falcon 2052 tubes, incubated with 4 ml of 1% BSA for 10 minutes, and centrifuged at 350g for 5 minutes. For determination of the percentage of NKT lymphocytes, anti-CD3 and anti DX5 antibodies are used (Pharmingen, USA). Analytical cell sorting is performed on 1x10 4 cells from each group with a fluorescence-activated cell sorter (FACSTAE plus, Becton Dickinson). Only live cells are counted, and background fluorescence from non-antibody-treated lymphocytes is subtracted. Gates are set on forward- and side-scatters to exclude dead cells and red blood cells. Data is analyzed with the Consort 30 two-color contour plot program (Becton Dickinson, Oxnard, CA), or the CELLQuest 25 program.
  • NKT lymphocytes Isolation of NKT lymphocytes. Cell separation is performed using Magnetic Cell Sorting (MACS, Miltenyi Biotec, Germany) according to the manufacturer's instructions. Anti-CD 3 and anti-DX5 magnetic beads are used for separation of NKT lymphocytes. Beads are removed between the two steps according to the manufacturer's instructions. Above 95% accuracy is achieved by FACS analysis of cells.
  • AST serum aspartic transaminase
  • ALT alanine aminotransferase
  • Blood glucose was measured with a standard glucometer.
  • Plasma triglyceride and total cholesterol values were measured by a clinical chemistry analyzer Reflovet Plus machine (Roche Diagnostics, GmbH, Mannheim, Germany).
  • Glucose Tolerance Test Mice were subjected to a glucose tolerance test (GTT) on day 30 after overnight fasting. Glucose was administered orally (1.25 g per kg). Serum glucose measurements were performed on tail-vein blood every 15 minutes for 3 hours. Glucose levels were measured by a standard glucometer.
  • GTT glucose tolerance test
  • IFN- ⁇ and TGF- ⁇ levels were determined on serum by "sandwich” ELISA, using commercial kits (Quantikine, R&D Systems, Minneapolis, MN, USA). Serum insulin was also determined by "sandwich” ELISA, using the commercial kit of Mercodia AB (Uppsala, Sweden) according to the manufacturer's instructions.
  • Triglyceride measurement On the day indicated, serum triglyceride levels are measured using a spectrophotometer (Cobas DP-25P).
  • Liver steatohepatitis score A liver segment from each mouse was fixed in 10% formaldehyde and embedded in paraffin for histological analysis. Five sections (5 ⁇ m) are stained with hematoxylin/eosin and reviewed by two pathologists in a blinded fashion. Histological examination and the steatohepatitis grade scoring (NASH score) are performed using the steatohepatitis scoring system.
  • Example 4 Oral administration of anti-LPS enriched colostrum-derived immunoglobulin preparation decreases liver enzymes
  • liver enzyme levels which indicate liver injury, of animals orally administered with anti-LPS enriched colostrum-derived immunoglobulin preparation are improved due to the treatment.
  • Levels of aspartyl transaminase (AST) and alanine aminotransferase (ALT) activities were determined by a clinical chemistry analyzer, Reflovet Plus (Roche Diagnostics, GmbH, Mannheim, Germany).
  • Figure 1 demonstrates the decrease was significant for AST group A versus B (* p ⁇ 0.001). This demonstrates amelioration of liver injury, as manifested by a clear and significant decrease in ALT and AST serum levels vs. the control group.
  • Example 5 Oral anti-LPS enriched colostrum-derived immunoglobulin preparation increases the expression of CD4+CD25+ regulatory T cells in the liver
  • Intrahepatic lymphocytes were isolated after mice were sacrificed, as follows: After the removal of livers, they were kept in medium (RPMI-1640+FCS). Then, livers were crushed through a stainless mesh (size 60, Sigma) and the cell suspension was placed in a 50-ml tube for 5min. Lymphoprep (10ml, Ficoll, Axis-Shield PoC AS, Oslo, Norway) was placed under similar volume of cell suspension in 50-ml tubes. Tubes were centrifuged at 1800 rpm for 18min.
  • Cells in the interface were collected and centrifuged at 1800 rpm for 10min, to obtain a pellet of cells depleted of hepatocytes to a final volume of 250ul. Approximately 1x10 6 cells/mouse liver, were recovered and analyzed by flow cytometry.
  • FIG. 1 To determine the specificity of the increase in regulatory T cells in the liver, the average surface expression of markers (CD4+CD25+) on hepatic lymphocytes was measured using flow cytometry on day 7 (sacrifice day) in all mice treated with 3.0 mg anti-LPS enriched colostrum-derived immunoglobulin preparation.
  • Figure 2A demonstrates oral anti-LPS enriched colostrum-derived immunoglobulin preparation increases the expression of CD4+CD25+ regulatory T cells in the liver. Gating was on CD4 and values are mean ⁇ SD.
  • FIG. 2B A representative dot blot derived from FACS performed on lymphocytes isolated from the livers of mice treated with anti-LPS enriched colostrum-derived immunoglobulin preparation (group B) or from untreated controls (group A) is shown in Figure 2B which shows oral anti-LPS enriched colostrum-derived immunoglobulin preparation increases the expression of CD4+CD25+ regulatory T cells in the liver.
  • Example 6 Oral anti-LPS enriched colostrum-derived immunoglobulin preparation increases the expression of CD25+CD4+LAP-, CD45+LAP+ and CD3+LAP+ regulatory T cells in the liver
  • LAP staining the following antibodies were used: anti-CD3-Alexa-fluor 405, anti-CD45-PerCP-Cy5.5 and anti -LAP-APC.
  • Affinity-purified biotinylated goat anti-LAP specific polyclonal antibody was purchased from R&D Systems (Minneapolis, MN, USA), and strepavidin-APC was used as secondary reagent for detecting the biotinylated primary antibody (R&D).
  • R&D biotinylated primary antibody
  • LAP staining cells were preincubated with LAP/control antibody for 20 min, and stained with CD3- Alexa-fluor 405 or CD45-PerCP-Cy5.5, followed by strepavidin-APC staining.
  • Figure 3 shows the average surface expression of markers (CD25+CD4+LAP-, CD45+LAP+ and CD3+LAP+) on hepatic lymphocytes measured using flow cytometry on day 7 (sacrifice day) in all mice treated with 3.0 mg. Values are means.
  • Figure 3A and B demonstrate oral administration of anti-LPS enriched colostrum-derived immunoglobulin preparation increased a subset of CD25+CD4+LAP-, CD45+LAP+ and CD3+LAP+ regulatory T cells in the liver.
  • Example 7 Oral anti-LPS enriched colostrum-derived immunoglobulin preparation increases the expression of CD45+LAP+ and CD8+LAP+ regulatory T cells in the spleen.
  • Figure 4 shows the average surface expression of markers (CD45+LAP+ and CD8+LAP+) on splenic lymphocytes measured using flow cytometry on day 7 (sacrifice day) in all mice treated with 3.0 mg. Values are mean ⁇ SD.
  • Figure 4A and B demonstrate oral administration of anti-LPS enriched colostrum-derived immunoglobulin preparation increases a subset of CD45+LAP+ and CD8+LAP+ regulatory T cells in the spleen.
  • Example 8 Oral administration of colostrum enriched with anti LPS antibodies to Ob/Ob mice
  • Ob/Ob model we used young (age 6-7 weeks) male C57BL/6 Ob/Ob mice which were purchased from Harlan Laboratories (USA). All mice were maintained in the Animal Core of the Hadassah-Hebrew University Medical School. Mice were administered standard laboratory chow and water ad libitum and kept in a 12-hour light/dark cycle. The animal experiments were carried out according to the guidelines of the Hebrew University-Hadassah Institutional Committee for Care and Use of Laboratory Animals and with the committee's approval.
  • mice of groups A-C following four weeks of T-IgG or HIBC administered orally.
  • Serum insulin was determined by "sandwich" ELISA, using the commercial kit of Mercodia AB (Uppsala, Sweden) according to the manufacturer's instructions.
  • Sera were collected from Ob/Ob mice on day 30 after sacrificing the mice.
  • Figure 5 demonstrates mice administered T-IgG exhibited a decrease in serum insulin levels, indicating the beneficial impact of the anti LPS antibodies on insulin resistance. Moreover, the decrease observed in provides data in support of an important role for the colostrum derived adjuvants in the metabolic effect.
  • Example 10 Oral anti-LPS enriched colostrum-derived immunoglobulin preparation decreases glucose tolerance in Ob/Ob mice.
  • Ob/Ob mice underwent a glucose tolerance test (GTT) on day 30 after overnight fasting. Glucose was administered orally (1.25g per kg). Serum glucose measurements were performed on tail-vein blood every 15 min for 3h. Glucose levels were measured by a standard glucometer.
  • GTT glucose tolerance test
  • mice administered HIBC improved glucose tolerance demonstrated by lower glucose values at glucose tolerance test with a decrease in the area under the curve as compared to the control group .
  • the data obtained in Examples 9 and 10 supports the importance of HIBC according to the present invention in the improvement of the metabolic syndrome.
  • Example 11 Oral administration of anti-LPS enriched colostrum-derived immunoglobulin preparation decreases liver injury in Ob/Ob mice.
  • liver enzyme levels which indicate liver injury, of animals fed with the preparation have also improved due to the treatment.
  • Levels of AST and ALT activities were determined by a clinical chemistry analyzer, Reflovet Plus (Roche Diagnostics, GmbH, Mannheim, Germany).
  • Figure 7 demonstrates a decrease of AST and ALT levels in T-IgG-colostrum-treated mice.
  • Example 12 Oral administration of anti-LPS enriched colostrum-derived immunoglobulin preparation decreases hepatic TGs in Ob/Ob mice.
  • Figure 8 demonstrates that oral administration anti-LPS enriched colostrum-derived immunoglobulin preparation decreased hepatic triglyceride content compared to mice in the control group. The decrease was significant for HIBC relative to controls (* P ⁇ 0.04).
  • Example 13 Oral administration of anti-LPS enriched colostrum-derived immunoglobulin preparation increases the expression of CD3+LAP+ regulatory T cells in the spleen.
  • Figure 9 shows the average surface expression of markers (CD3+LAP+) on splenic lymphocytes measured using flow cytometry on day 25 (sacrifice day) in all Ob/Ob mice. Values are mean ⁇ SD.
  • Figure 9A and B demonstrate oral administration of T-IgG increases a subset of CD3+LAP+ regulatory T cells in the spleen.
  • Example 14 Oral administration of anti-LPS enriched colostrum-derived immunoglobulin preparation increases the expression of CD8+CD25+ regulatory T cells in the spleen
  • Figures 10 and 11 demonstrate oral administration of T-IgG increases a subset of CD8+CD25+ regulatory T cells in the spleen.
  • Example 15 Oral anti-LPS enriched colostrum-derived immunoglobulin preparation increases the expression of CD4+CD25+ regulatory T cells in adipose tissue
  • FIGS. 12A and 12B show the average surface expression of markers (CD4+CD25+) on adipose tissue lymphocytes measured using flow cytometry on day 25 (sacrifice day) in all Ob/Ob mice.
  • Figure 12 demonstrates oral administration of T-IgG increases a subset of CD4+CD25+ regulatory T cells in adipose tissue.
  • Example 16 Oral anti-LPS enriched colostrum-derived immunoglobulin preparation increases the expression of CD3+LAP+ regulatory T cells in adipose tissue
  • FIGS. 13A and 13B show the average surface expression of markers (CD3+LAP+) on adipose tissue lymphocytes measured using flow cytometry on day 25 (sacrifice day) in all Ob/Ob mice.
  • Figure 13 demonstrates oral administration of T-IgG increases a subset of CD3+LAP+ regulatory T cells in adipose tissue.
  • Example 17 Oral anti-LPS enriched colostrum-derived immunoglobulin preparation increases the expression of CD4+CD25+ regulatory T cells in Stromal Vascular Cells (containing preadipocytes)
  • Figures 14A and 14B show the average surface expression of markers (CD4+CD25+) on adipose tissue lymphocytes measured using flow cytometry on day 25 (sacrifice day) in all Ob/Ob mice.
  • Figure 14 demonstrates oral administration of T-IgG increases a subset of CD4+CD25+ regulatory T cells in the Stromal Vascular Cells containing preadipocytes.
  • Figures 15A and 15B show the average surface expression of markers (CD4+CD25+LAP+) on adipose tissue lymphocytes measured using flow cytometry on day 25 (sacrifice day) in all Ob/Ob mice.
  • Figure 15 demonstrates oral administration of T-IgG increases a subset of CD4+CD25+ regulatory T cells in the Stromal Vascular Cells containing preadipocytes.
  • Example 18 Dosage studies of anti-LPS enriched colostrum-derived immunoglobulin preparation in Ob/Ob mice
  • mice were sacrificed. On sacrifice day, cardiac blood was collected by standard techniques then serum was obtained for future purposes.
  • Ob/Ob model we used young (age 6-7 weeks) male C57BL/6 Ob/Ob mice which were purchased from Harlan Laboratories (USA). All mice were maintained in the Animal Core of the Hadassah-Hebrew University Medical School. Mice were administered standard laboratory chow and water ad libitum and kept in a 12-hour light/dark cycle. The animal experiments were carried out according to the guidelines of the Hebrew University-Hadassah Institutional Committee for Care and Use of Laboratory Animals and with the committee's approval.
  • Example 19 Oral administration of anti-LPS enriched colostrum-derived immunoglobulin decreases liver enzymes in Ob/Ob mice.
  • Example 20 Oral administration of anti-LPS enriched colostrum-derived immunoglobulin decreases total cholesterol in Ob/Ob mice.
  • Plasma triglycerides and total cholesterol were determined by a clinical chemistry analyzer, Reflovet Plus (Roche Diagnostics, GmbH, Mannheim, Germany) as described above.
  • Figure17 demonstrates 100ug of T-IgG was the most effective dose in decreasing total cholesterol.
  • Example 21 Oral administration of anti-LPS enriched colostrum-derived immunoglobulin decreases hepatic TGs in Ob/Ob mice.
  • TGs intracellular triglycerides
  • Figure18 demonstrates 100ug of 1mg, 3mg and 100ug of T-IgG were the most effective doses in decreasing hepatic triglycerides. The decrease was statistically significant for group A versus D, E, F (* p ⁇ 0.05).
  • Example 22 Oral administration of 1ug, 1mg, 3mg of T-IgG, along with 100ug HIBC, decreased CD3+NK1.1+ cells in the livers of Ob/Ob mice.
  • FACS analysis was performed on lymphocytes isolated from livers of Ob/Ob mice. Average of expression of markers (CD3+NK1.1+) on hepatic lymphocytes was measured using flow cytometry on day 25 (sacrifice day) in all ob/ob mice. For flow cytometry, the following antibodies were used: anti-CD3-FITC and anti NK1.1- PE. Surface staining and FACS analysis was performed as described above.
  • Figure 19A demonstrates oral administration of 1ug, 1mg, 3mg of T-IgG, along with 100ug HIBC, decreased CD3+NK1.1+ cells in the livers of Ob/Ob mice.
  • Figure 19B demonstrates oral administration of 1ug and 100ug of T-IgG, decreased CD3+NK1.1+ cells in the livers of Ob/Ob mice
  • Example 23 Oral administration of T-IgG and HIBC colostrums, increases CD4+CD25+LAP-/LAP+ cells in the livers of Ob/Ob mice
  • Figure 20A demonstrates oral administration of T-IgG and HIBC colostrums, increases CD4+CD25+LAP-/LAP+ cells in the livers of Ob/Ob mice.
  • Figure 20B demonstrates oral administration of 100ug of HIBC colostrum, increases CD4+CD25+LAP+ cells in the livers of Ob/Ob mice.
  • Example 24 Oral administration of T-IgG and of HIBC-colostrums, induces changes in CD25+LAP- hepatic lymphocytes
  • Figure 21 demonstrates oral administration of 1ug, 1mg, 3mg of T-IgG, along with 100ug HIBC, induces changes in CD25+LAP- lymphocytes in the livers of Ob/Ob mice.
  • Example 25 Oral administration of T-IgG and of HIBC-colostrums induces changes in CD25+LAP+ splenic lymphocytes
  • Figure 22A demonstrates administration of T-IgG and of HIBC-colostrums, decreases CD25+LAP+ splenic lymphocytes.
  • Figure 22B demonstrates oral administration of T-IgG-colostrums increases CD25+LAP+ splenic lymphocytes.
  • Example 26 Oral administration of 1 and 3mg of T-IgG and of 100ug of HIBC-colostrums, increases CD4+CD25+LAP- splenic lymphocytes
  • Figure 23 shows the average of expression of markers (CD4+CD25+LAP-) on splenic lymphocytes was measured using flow cytometry on day 25 (sacrifice day) in all ob/ob mice.
  • Figure 23 demonstrates oral administration of 1 and 3mg of T-IgG and of 100ug of HIBC-colostrums, increases CD4+CD25+LAP- splenic lymphocytes
  • Example 27 Oral administration of T-IgG -colostrums, increases CD4+CD25+ in adipose tissue.
  • Figure 24 shows Average of expression of markers (CD4+CD25+) on adipose tissue cells was measured using flow cytometry on day 25 (sacrifice day) in all ob/ob mice.
  • Figure 24 demonstrates oral administration of T-IgG-colostrums, increases CD4+CD25+ lymphocytes in adipose tissue.
  • Example 28 Oral administration of 100ug of T-IgG-colostrum, increases CD4+CD25+ in adipocytes.
  • Figure 25A demonstrates the average of expression of markers (CD4+CD25+) on adipocytes was measured using flow cytometry on day 25 (sacrifice day) in all ob/ob mice.
  • Figure 25A demonstrates administration of 100ug of T-IgG -colostrum, increases CD4+CD25+ in adipocytes.
  • Figure 25B demonstrates oral administration of 100ug of T-IgG -colostrum, increases CD4+CD25+ in adipocytes.
  • Example 29 Oral administration of T-IgG-colostrum, increases CD3+LAP+ in adipocytes
  • FIG. 26 shows the average of expression of markers (CD3+LAP+) on adipocytes was measured using flow cytometry on day 25 (sacrifice day) in all ob/ob mice.
  • Figure 26A demonstrates oral administration of T-IgG -colostrum, increases CD3+LAP+ in adipocytes.
  • Figure 26B demonstrates oral administration of T-IgG-colostrum, increases CD3+LAP+ in adipocytes.
  • Example 30 Oral administration of T-IgG -colostrum, increases CD4+CD25+LAP- in adipocytes
  • Figure 27A demonstrates administration of T-IgG-colostrum, increases CD4+CD25+LAP- in adipocytes.
  • Example 31 Oral administration of anti LPS enriched colostrum decreases bacterial translocation in a model of hepatitis
  • mice were treated as follows: Group A: Treated with BCP antibody free colostrum; Group B: anti LPS containing colostrum. Mice were fed with colostrum for 4 days prior to induction of Con A hepatitis.
  • Con A was purchased from MP Biomedicals (Ohio, USA). Con A (0.5 mg, 20 mg/kg) was dissolved in 200 uL of 50 mM Tris (pH 7), 150 mM NaCl, 4 mM CaCl 2 , and injected intravenously into mice. Sera from individual mice were obtained 8 or 20 h after Con A injection. Serum activities of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were measured using an automatic analyzer.
  • ALT alanine aminotransferase
  • AST aspartate aminotransferase
  • LPS lipopolysaccharide
  • LAL limulus amebocyte lysate
  • Group A A1 28170 ConA+BCP A2 857.6 A3 1356.8 A4 340.8 A5 26340 11413 . 04 14480.59
  • Group B B1 10992 ConA + T-IgG B2 796.8 B3 187.2 B4 2816 B5 12672 5492 . 8 5898.076
  • Example 32 Preparation of compositions containing colostrum-derived anti-LPS enriched preparations and anti-insulin antibodies
  • colostrum was collected from approximately 200 commercial dairy cowherds. The cows in these herds, as well as being vaccinated for routine cattle pathogens, have been vaccinated with a vaccine against the outer cell wall antigens of multiple strains of E. coli bacteria, a major organism in human gut microflora.
  • colostrum was collected from approximately 200 commercial dairy cowherds. The cows in these herds, as well as being vaccinated for routine cattle pathogens, have been vaccinated with a vaccine against the outer cell wall antigens of multiple strains of E. coli bacteria, a major organism in human gut microflora.
  • three dairy cows are immunized with insulin conjugated to KLH as an antigen.
  • the antigen vaccines are administered during the last eight weeks of gestation.
  • Colostral milk is collected during the first two days of lactation. The obtained colostrum was frozen in individual bags for testing.
  • colostrum was thawed, pooled and fat was removed. Each batch was pasteurized. Colostrum was concentrated by ultra-filtration to reduce volume before freeze drying. The ultra-filtration step reduced lactose in the final powder to less than 7% (from about 50%).
  • the anti-LPS enriched immunoglobulin preparation and the anti-insulin enriched immunoglobulin preparation are mixed to form a composition for use as described below.
  • eleven to twelve weeks old male C57/bl mice are tail vein injected with a dose of 500 ⁇ g linouse (approximately 15 mg/kg) of Con A (MP Biomedicals, USA) which is dissolved in 50mM Trig pH 7, 150mM NaCl, 4mM CaCl 2 , known to induce hepatitis. Animals of all tested groups are orally administered (e.g.
  • AST serum aspartate aminotransferase
  • ALT alanine aminotransferase
  • histological examination of liver specimens FACS analysis of intrahepatic and intrasplenic lymphocytes for NKT markers, measurement of TGs, total cholesterol, glucose tolerance, serum insulin, serum glucose, cytokine levels and Western blot analysis for the expression of the transcription factors STAT 1, 4 and 6 and NF ⁇ B and are compared to control groups.
  • anti-LPS immunoglobulin preparation can exert an immunomodulatory effect and alleviate target organ damage in animal models.
  • a study was performed to determine the safety and efficacy of oral administration of anti-LPS immunoglobulin preparation to patients with insulin resistance and NASH.
  • CBCP control BCP
  • HIBC preparations anti-insulin immunoglobulin preparation
  • Candidates were identified from among the patients treated in the department of medicine and liver unit of the Hadassah Hebrew University Medical Center and signed an approved informed consent from before the study activities were initiated. Participants were be randomized to receive either control bovine (CBCP) (30 patients) or anti-LPS immunoglobulin preparation (20 patients). Patients were followed on study with weekly visits as well as for an additional 4 weeks after concluding treatment to assess safety.
  • CBCP control bovine
  • HIBC preparations anti-insulin immunoglobulin preparation
  • control BCP 1.8 grams (3 x 200 mg capsules), anti-LPS immunoglobulin preparation (HIBC) 1.8 g daily (3 x 200mg capsules, three times daily) or anti-insulin immunoglobulin preparation 600 mg per day (2 x 100 mg capsules, three times daily), for 4 weeks and then monitored off study treatment for an additional 4 weeks.
  • HIBC anti-LPS immunoglobulin preparation
  • each 200 mg tablet contains 200 mg of freeze-dried BCP from cows immunized with LPS as the only active component in combination with excipients including silica colloidal anhydrous, magnesium stearate, microcrystalline cellulose and calcium carbonate.
  • the product can be stored at room temperature and has a shelf life of 5 years.
  • each 100 mg tablet contains 100 mg of freeze-dried BCP from cows immunized with insulin as the only active component in combination with excipients including silica colloidal anhydrous, magnesium stearate, microcrystalline cellulose and calcium carbonate.
  • the product can be stored at room temperature and has a shelf life of 5 years.
  • compositions were stored and dispensed by the research pharmacies at the Hadassah Hebrew University Medical Center.
  • the site pharmacist is required to maintain complete records of all study products.
  • concomitant medications there are no specific protocol-imposed restrictions on concomitant medications, other than stipulated in the inclusion/exclusion criteria. Nonetheless, whenever a concomitant medication or study agent is initiated or a dose changed, investigators reviewed the concomitant medications' and study agents' most recent package inserts, investigator's brochures, or updated information from on-line sources to obtain the most current information on drug interactions, contraindications, and precautions.
  • Each candidate subject was scheduled for a screening visit.
  • the study investigator(s) explained the study in detail, answered any questions the candidate may have had, and gave the candidate a consent form to read and sign.
  • the candidate subject was asked to provide a complete medical history and undergo a physical examination, including measurement of blood pressure, pulse, temperature, body weight and height.
  • CBC complete blood count
  • Other laboratory analyses See Table 6
  • Serum was collected and archived for use in the development of surrogate markers. Women of childbearing potential underwent a pregnancy test. The subjects' medication history was evaluated to determine that both the medication itself and the dose regimen of the medication fall within the inclusion criteria.
  • CBCP anti-LPS immunoglobulin preparation or anti-insulin immunoglobulin preparation
  • Results of routine diagnostic tests obtained during pretreatment evaluation or during the course of research project will be made available to the subject's primary care physician upon receipt by the principal investigator of a signed written consent from the candidate subject.
  • the CBCP, anti-LPS immunoglobulin preparation, and anti-insulin immunoglobulin preparation were taken every day for 30 days.
  • CBCP CBCP or immunoglobulin preparations
  • a subject forgets to take the drug in the morning, he/she can take the drug during the day. He/she is required not to ingest food for 2 hours before and 2 hours after taking the drug. Subjects were treated on an outpatient basis and were monitored during a 30 day, treatment period and a 30 days follow-up after completion of the study.
  • Visit 1 will took place up to 14 days after the screening visit. In case of a delay, the screening visit was repeated.
  • the subject Prior to engaging in any study procedures, the subject must meet the inclusion/exclusion criteria by history (which includes a signed declination), and review and sign an informed consent form.
  • Visit 2-5 Days 7,14, 21, 30
  • hematocrit mean corpuscular volume (MCV)
  • WBC white blood cell count
  • ANC absolute neutrophil count
  • Activated T cells defined as those coexpressing CD38 and HLA-DR.
  • the medical history included, at a minimum, duration of known HCV infection, previous hospitalizations and diagnoses, with a special emphasis on liver related complications and other HCV-related conditions, exposure to antiviral medications, and any pre-existing medical conditions that may interfere with the conduction of the study.
  • a complete physical examination is to include at a minimum an examination of the skin, head, mouth, and neck; auscultation of the chest; cardiac exam; abdominal exam; examination of the lower extremities for edema.
  • the complete physical exam also includes signs and symptoms, diagnoses, and vital signs (temperature, pulse, respiration rate, and blood pressure).
  • a targeted physical examination includes vital signs (temperature, pulse, respiration rate, and blood pressure) and is to be driven by any previously identified or new symptoms that the subject has experienced or diagnoses that have been made on the subject since the last visit.
  • Women who become pregnant during the study will discontinue study treatment, i.e. will not receive any further doses of colostrum, and report pregnancy-related information (e.g., complications, births, fetal loss/abnormalities). These subjects were encouraged to remain in the study to be followed off study treatment/on study, for safety evaluations (clinical assessment, targeted exam) per protocol until study completion and will be followed by telephone contact thereafter to determine the pregnancy outcome. Outcomes (health of the infant) and any pregnancy-related complications must also be recorded.
  • pregnancy-related information e.g., complications, births, fetal loss/abnormalities.
  • the baseline value (defined as the average of 2 observations prior to study treatment) was compared to each of the on-treatment measurements and each of the post-treatment observations, and the values at the end of the treatment period to the post-treatment values.
  • a similar approach was used to test the secondary hypothesis of a decrease in immune activation after the administration of study treatment.
  • CRF RECORDS TO BE KEPT Case report forms
  • Example 34 Alleviation of insulin resistance and liver damage by oral administration of Anti-LPS immunoglobulin enriched colostrum preparation is mediated by increased Tregs and associated with increased serum GLP-1 and adiponectin: Results of a Phase I/II clinical trial in NASH
  • NASH Inclusion criteria. Ten patients were enrolled in an open-label trial. Participants (men and women between 18 and 60 years old) were evaluated for eligibility after they signed a written informed consent form. The diagnosis of NASH was based on a liver biopsy score of 4 or above and altered glucose metabolism, including diabetes (non-treated or treated with up to two drugs, without any change in medication two months prior to enrollment), impaired fasting glucose or impaired glucose tolerance and HbA1C between 5.5 and 14%. Impaired fasting glucose was defined as >100 mg/dl. Impaired glucose tolerance was defined as a blood sugar level >140 mg/dl two hours post-glucose load and an HBA1C between 5.5 and 8%. No evidence of other viral or immune-mediated liver disease was present.
  • Exclusion criteria Patients meeting any of the following criteria were excluded: active co-infection with hepatitis A, B, or C viruses; the presence of human immunodeficiency virus (HIV) infection, hepatocellular carcinoma, fulminant liver failure, severe deteriorating synthetic liver functions or a clinically significant infectious, immune-mediated or malignant disease; any history of treatment with immunomodulatory drugs, including steroids and NSAID, at any time within the last four weeks; a history of coagulopathy; women with childbearing potential unless surgically sterile or using adequate contraception (i.e., IUD, oral or Depo-Provera contraceptive or barrier plus spermicide); anemia (Hb ⁇ 10.5 gm/dl), thrombocytopenia (platelets ⁇ 100 k/ ⁇ l) or lymphopenia (absolute lymphocyte count ⁇ 0.7) or Allergy to cow milk or lactose intolerant.
  • HBV human immunodeficiency virus
  • CBC complete blood counts
  • ESR sedimentation rate
  • standard chemistries including liver enzymes, INR, lipid profile, CRP, HbA1C, and serum insulin level. All patients underwent a repeat oral glucose tolerance test and a HOMA score evaluation at the end of the study.
  • Treated patients showed a decrease in serum levels of triglycerides (1.88 vs. 1.32 ⁇ mol/L, p ⁇ 0.05), total cholesterol (5.28 vs. 4.44 ⁇ mol/L, p ⁇ 0.04), and LDL. cholesterol (3.7 vs. 2.49 ⁇ mol/L, p ⁇ 0.05).
  • a decrease in liver enzymes was noted in most treated patients (ALT: 54.5 vs. 43.16, u/l, p ⁇ 0.04; AST: 50.58 vs. 45.5 u/l, p ⁇ 0.05; Alkaline phosphatase: 82.1 vs. 72.4 u/l, p ⁇ 0.001; GGT: 84.3 vs.
  • Example 35 Oral administration of anti-LPS immunoglobulin preparation decreases liver injury in humans.
  • liver enzymes AST and ALT, AP and GGT
  • serum fasting glucose serum fasting glucose
  • insulin plasma lipids
  • plasma lipids cholesterol and triglycerides
  • HIBC anti-LPS immunoglobulin preparation
  • Figures 28A-B, 29, 30 and 31 show the effect of oral administration of anti-LPS immunoglobulin preparation (HIBC) at a dose of 1.8 g/day on serum levels of ALT, AST, Alkaline phosphatase (AP) and ⁇ -GGT, respectively.
  • ALT levels were decreased in five patients (57.4 vs. 48.6u/L, for day 1 vs. day 30; P ⁇ 0.04).
  • AP levels were improved in eight treated patients (83.1 vs. 73.9u/L, for day 1 vs. day 30; P ⁇ 0.002) and ⁇ -GGT levels were decreased in five treated patients (88.2 vs. 73.2u/L, for day 1 vs. day 30; P ⁇ 0.05).
  • the methods can include identifying a subject with a level of ALT above a given level, e.g., 50 U/L, and administering the anti-LPS enriched colostrum as described herein.
  • the methods can include identifying a subject with a level of AST above a given level, e.g., 50 U/L, and administering the anti-LPS enriched colostrum as described herein.
  • the methods can include identifying a subject with a level of AP above a given level, e.g., 70 U/L, and administering the anti-LPS enriched colostrum as described herein.
  • the methods can include identifying a subject with a level of GGT above a given level, e.g., 60 U/L, and administering the anti-LPS enriched colostrum as described herein.
  • Example 36 Oral administration of anti-LPS immunoglobulin preparation decreases fasting glucose levels, increases early peak insulin secretion, improves oral glucose tolerance and improves HB1Ac levels, HOMA scores, GLP-1 levels in humans
  • Figures 35A-B show a significant improvement in HbAlc values in all treated patients (7.49 vs. 6.38%, for day 1 vs. day 30, respectively, for ten patients; P ⁇ 0.001).
  • the treatment with an anti-LPS immunoglobulin enriched colostrum preparation for 30 days caused a 14.8% decrease in HbAlc values in all ten patients.
  • the greatest decrease occurred in patients with higher baseline HbAlc values.
  • the methods can include identifying a subject with a level of HbAlc above a given level, e.g., over 7 or 8, and administering the anti-LPS enriched colostrum as described herein.
  • a threshold level e.g., above 300 pg/ml
  • Figure 36A-B show the improved HOMA scores (6.7 vs. 4.8, for day 1 vs. day 30, respectively, for six patients).
  • the methods can include selecting subjects who have a HOMA score above a threshold, e.g., above 7 or 8, and administering the anti-LPS enriched colostrum as described herein.
  • GLP-1 The central role of GLP-1 in glucose tolerance has raised questions about the possible involvement of this peptide in the pathogenesis of diabetes.
  • circulating cytokines and adiponectin were made as follows. Blood was drawn from all patients on day 1 and on day 30 of the study. Serum levels of IL-6 were determined using a "sandwich” ELISA using commercial kits (Quantikine, R&D Systems, Minneapolis, MN, USA), according to the manufacturer's instructions. Glucagon-Like Peptide-1 (GLP-1) was tested by the following method: Blood was collected from all patients after a 12-hour overnight fasting at 180 min time point of OGTT. Blood was collected in ice-cooled EDTA tubes and immediately ( ⁇ 30 seconds) after collection, 20 ⁇ l of DPP-IV inhibitor was added to 2ml of plasma, according to manufacturer's directions.
  • GLP-1 Glucagon-Like Peptide-1
  • Tubes were inverted and were then centrifuged immediately at 1000 x g for 10 min in refrigerated centrifuge. After plasma separation, specimens were stored at -70°C until running the ELISA assay.
  • the circulating level of human GLP-1 was quantified using a commercial ELISA kit from Millipore (MA, USA) according to the manufacturer's instructions. Serum levels of human adiponectin were determined using a commercial ELISA kit from Linco Research (Missouri, USA).
  • Serum was diluted 500 fold before the assay, and 20 ⁇ l of diluted serum, standard samples and controls were plated in duplicate on a mouse anti-human adiponectin-coated plate and examined using an ELISA reader at 450 nm, according to the manufacturer's instructions.
  • FIG. 37A-B show that treatment for 30 days increased serum levels of GLP-1 post-OGTT in five treated patients (6.31 vs. 6.78 X 10 4 pM, in responders).
  • Figures 38A-B show the serum levels of adiponectin, which were increased in eight patients (6181 vs. 7069 ng/ml).
  • oral anti-LPS immunoglobulin preparation at a dose of 1.8 g/day increases GLP-1 levels and adiponectin1 levels in humans.
  • Example 37 Oral administration of anti-LPS immunoglobulin preparation decreases total cholesterol levels, LDL cholesterol levels and triglyceride levels in humans
  • Figures 39A-B show a beneficial effect of the anti-LPS immunoglobulin preparation on serum levels of total cholesterol in five treated patients (5.8 vs. 4.5 ⁇ mol/L, for day 1 vs. day 30, respectively). Similar results were obtained with serum LDL measurements (3.8 vs. 2.7 ⁇ mol/L, for day 1 vs. day 30, respectively, Figures 40A-B ). A slight improvement in serum triglycerides levels was noted in five patients ( Figures 41A-B ).
  • the methods can include selecting subjects who have a level of total serum cholesterol above a threshold, e.g., above 6 uM/dL; a level of serum LDL above a threshold, e.g., 4 uM/dL, and/or a level of serum triglycerides above a threshold, e.g., above 2 or 2.5 uM/dL, and administering the anti-LPS enriched colostrum as described herein.
  • a threshold e.g., above 6 uM/dL
  • a level of serum LDL above a threshold e.g., 4 uM/dL
  • a level of serum triglycerides above a threshold e.g., above 2 or 2.5 uM/dL
  • Example 38 Oral administration of anti-LPS immunoglobulin preparation decreases increases CD4+ CD25+ and CD4+ CD25+ Foxp3+ T regulatory cells in humans
  • PBMCs peripheral T cells from PBMCs were characterized using flow cytometry. PBMCs from blood samples collected at day 0 and day 30 were isolated using a Ficoll-Hypaque gradient. Cells were re-suspended in PBS containing 1% BSA.
  • PBMCs were incubated with either fluorochrome-conjugated antibodies against the indicated cell surface markers (eBioscience, San Diego, CA, USA) at the recommended dilution or with isotype control antibodies for 30 minutes at 4°C.
  • cell surface antibodies CD4-FITC, CD25-PE, CD8-FITC, CD56-FITC, CD69-PE, CD3-APC, CD62-PE and HLA-DR-APC.
  • Cells were then washed in PBS containing 1% BSA and fixed with fixation buffer (eBiosciences) for another 50 minutes.
  • Figures 42A-B show that CD4+CD25+ cells were elevated 30 days after oral treatment with an anti-LPS immunoglobulin enriched colostrum preparation in 7 out of the 10 patients (4.63 vs. 6.28%).
  • Figure 42D shows an increase in CD4+CD62+ cells which was noted in six patients (36.5 vs. 41 % in responders, gated for CD4 cells).
  • Example 40 Oral administration of anti-LPS immunoglobulin preparation in mice
  • the metabolic syndrome is a chronic inflammatory condition.
  • Regulatory T cells are essential for maintaining peripheral tolerance and limiting chronic inflammatory diseases. Tregs were shown to alleviate the pathological and metabolic abnormalities accompanied NASH in ob/ob mice.
  • Aim To assess the effects of the induction of Tregs on hepatic injury and insulin resistance in NASH.
  • Leptin deficient Ob/Ob mice were fed for 6 weeks with bovine colostrum powder (BCP) prepared from cows that were not immunized or with colostrum from cows that were "hyper immunized” with LPS (Enterotoxigenic E.coli, 0.1mg per dose), or with IgG-enhanced fraction of anti-LPS immunoglobulin preparation ('T-IgG') in three dosages, 0.001, 0.1 and 1 mg.
  • BCP bovine colostrum powder
  • LPS Enterotoxigenic E.coli, 0.1mg per dose
  • 'T-IgG' IgG-enhanced fraction of anti-LPS immunoglobulin preparation
  • Hepatic injury and insulin resistance were measured by fasting glucose levels, glucose tolerance tests (GTT) and liver enzymes. Fat accumulation in the liver and plasma lipids were measured. Serum TNF-alpha was determined by ELISA and the staining of Tregs in the spleen and liver was performed
  • CD4+CD25+ cells P ⁇ 0.01, P ⁇ 0.05, for 0.001, 0.1 and 1mg anti-LPS immunoglobulin preparation, and T-IgG, respectively
  • CD4+CD25+Foxp3 cells P ⁇ 0.001, P ⁇ 0.05 for 0.001mg of anti-LPS immunoglobulin preparation, and 0.1mg of T-IgG, respectively
  • CD3+NK1.1 cells P ⁇ 0.05 for 0.1mg of anti-LPS immunoglobulin preparation, and 0.1mg of T-IgG.
  • Example 41 Oral administration of anti-insulin immunoglobulin preparation in humans
  • Metabolic syndrome is a chronic inflammatory disorder associated with insulin resistance and hepatic steatosis.
  • Anti insulin (AI) colostrum 'anti-insulin immunoglobulin preparation' as described herein
  • Aim To determine the safety and efficacy of oral administration of anti-insulin immunoglobulin preparation to patients with insulin resistance and NASH.
  • liver enzymes were noted in most treated patients (ALT: 67 vs. 48, u/L, p ⁇ 0.01; AST: 59 vs. 40 u/L, p ⁇ 0.05; Alkaline phosphatase: 101 vs. 91 u/L, p ⁇ 0.004; GGT: 70 vs. 58. u/L, p ⁇ 0.004).
  • a weight loss of at least 15% of the original body weight was observed in 90% of treated subjects (80.5 vs.
  • Example 42 Oral administration of anti-insulin immunoglobulin preparation decreases liver injury in humans.
  • Figures 44, 45 , 46 and 47 demonstrate oral administration of anti-insulin immunoglobulin preparation (anti-insulin HIBC) at a dose of 1.2 g/day decreases liver injury in humans.
  • Example 43 Oral administration of anti-insulin immunoglobulin preparation decreases fasting glucose levels, increases early peak insulin secretion, improves oral glucose tolerance and improves HB1Ac levels, improves HOMA scores, GLP-1 levels in humans
  • Figure 48 demonstrates oral anti-insulin immunoglobulin preparation at a dose of 1.2 g/day decreases fasting glucose levels in humans.
  • Figure 49 demonstrates anti-insulin immunoglobulin preparation at a dose of 1.2 g/day increases early peak insulin secretion in humans.
  • Figure 50 demonstrates oral anti-insulin immunoglobulin preparation at a dose of 1.2 g/day improves oral glucose tolerance in humans.
  • Example 44 Oral administration of anti-insulin immunoglobulin preparation decreases total cholesterol levels, LDL cholesterol levels and triglyceride levels in humans
  • Figure 51 demonstrates oral anti-insulin immunoglobulin preparation at a dose of 1.2 g/day decreases total cholesterol levels in humans.
  • Figure 52 demonstrates anti-insulin immunoglobulin preparation at a dose of 1.2 g/day decreases body weight in humans.
  • Example 45 Oral administration of anti-insulin immunoglobulin preparation decreases increases CD4+ CD25+ T regulatory cells in humans
  • Figure 53 demonstrates oral administration of anti-insulin immunoglobulin preparation (HIBC) at a dose of 1.2 g/day results in increased CD4+CD25+ Tregs in 60% of patients (2.95% vs. 4.27%, p ⁇ 0.003).
  • HIBC anti-insulin immunoglobulin preparation
  • Example 46 Oral administration of anti-insulin immunoglobulin preparation decreases waist and arm circumference in humans
  • Figures 54 and 55 demonstrate oral administration of anti-insulin immunoglobulin preparation (HIBC) at a dose of 1.2g/day results in decreased waist and arm circumference in humans.
  • HIBC anti-insulin immunoglobulin preparation
  • Dairy cows at commercial dairy farms are immunized with either a killed single strain ETEC vaccine (078 serotype) or a killed multivalent ETEC vaccine in adjuvant (combined 06, 08, 015, 025, 027, 063, 0114, 0115, 0128, 0148, 0153 and 0159 serotypes).
  • the single strain vaccine is used to immunize approximately one third of the cows against one of the most common ETEC strains (serotype 078) and the multi-strain vaccine is used to immunize the other two-thirds of the cows in the immunization program.
  • Inoculation of vaccine plates - starter suspension is inoculated onto multiple CFA plates. These vaccine plates are incubated at 37°C for 18-24hrs under aerobic conditions.
  • a haemagglutination test is carried out on one of the vaccine plates to confirm pilus production.
  • Enumeration of Vaccine Washings The "vaccine washings" step is enumerated.
  • the material should be thoroughly agitated to disperse all clumps and ensure that the dilutions chosen are appropriate to the degree of concentration of bacterial cells in the washings for the batch being manufactured.
  • the "vaccine washing” is homogenized using a sterilized probe with a homogenizer for a total of 15 minutes at one minute intervals, with one minute of cooling in an ice-bath between each interval.
  • Pilus/LPS fraction Separation and dialysis of Pilus/LPS fraction -
  • the "homogenized vaccine washing” is centrifuged at 12,000 g for 20 minutes at 4°C and the supernatant is kept. Sterile saturated ammonium sulphate solution is added slowly to the supernatant until 20% saturation is reached. Recentrifuge the mixture and again keep the supernatant. Add further saturated ammonium sulphate solution until 40% saturation is reached and then recentrifuged again. The "Pilus/LPS” fraction is located in the pellet. The “Pilus/LPS” fraction is resuspended and then dialyzed, using a 3,500 MW cut-off membrane, for 24-48 hours at 4°C against 250-1,000 volumes of cold 0.05M sodium phosphate buffer pH 7.2.
  • Equal quantities of thawed "formalinized Pilus/LPS dialysate” and Seppic Montanide ISA 206 adjuvant are warmed to 30°C and then mixed together gradually while stirring. After overnight storage at 4°C, vaccine is re-mixed and then filled into sterile pillow packs.
  • Final QC of the vaccine before release includes testing for sterility, free formalin level and safety test for one week at 2 x normal dose in 3 cows.
  • the Immuron Multi Strain E.coli Vaccine is derived from the equivalent cell mass as the Single Strain E.coli Vaccine, with each strain contributing an equal share of the total.
  • Each of the 12 strains are prepared using the above process and then pooled before final formulation with adjuvant.
  • the 12 strains are shown in Table 7A.
  • Table 7A Enterotoxigenic E. coli Strains in Immuron's Killed ETEC Vaccines Serotype Strain no.
  • Source Date of Isolation (ref) ETEC O6: H16 B2C USA pre-1971 (1)
  • ETEC O8 H19 C55 3/3c3 USA mid-1980's
  • ETEC O15 H4 PE 595 IMVS, Sydney Aust.
  • Each 2ml of Single Strain E. coli Vaccine contains: 1ml of "Formalised Pilus /LPS Dialysate" from 078 E.coli serotype Prepared as described in Allied Biotech Doc. No. S04gs Final conc. of "Pilus/LPS" immunogen is min. of 1.0 mg/ml Immunogen - Containing 0.05M sodium phosphate buffer Ph 7.2 Buffer - Free Formalin ⁇ 0.002% w/v in final emulsion Inactivates vaccine Plus 1ml of Seppic Montanide ISA206 Adjuvant Adjuvant B. Each 2ml of Multi Strain E.
  • coli Vaccine contains: Immunogen 1ml of "Formalised Pilus /LPS Each strain contributing an equal share (1/12) of the total minimum equivalent cell mass needed to produce the 1.0mg/ml final concentration of the Immuron Single Strain E.coli vaccine Dialysate" from the following E.coli serotypes: 06, 08, 015, 025, 027, 063, 0114, 0115, 0128, 0148, O153 and 0159. Prepared as described in Allied Biotech Doc. No. S04gm - Containing 0.05M sodium phosphate buffer Ph 7.2 Buffer Inactivates - Free Formalin vaccine Plus 1ml of Seppic Montanide Adjuvant ISA206 Adjuvant
  • All immunizations are administered using clean handling procedures in order to maximize immune response to the antigens, and to minimize any reaction to the injection.
  • the selected cows are yarded and a sub-cutaneous injection is administered.
  • the cows are immunized a further 2 times leading up to calving at 2-4 week intervals, ceasing at least 1 month before calving.
  • Injections are given by the sub-cutaneous route after thoroughly disinfecting site by scrubbing with an iodine preparation.
  • a hygienic, food grade polyethylene bag is used to collect and store the colostrum at each farm.
  • the colostrum is filtered as it enters the bag using a 200 micron filter, to remove gross matter.
  • the colostrum is harvested from Holstein Friesian and Jersey cows. They are all government registered and at the time of harvesting are free from antibiotics. They are not given steroids at any stage of the process. Colostrum is harvested at the first milking which will be within twelve hours of calving. Each cow produces approximately 8 liters of colostrum. New born calves are given a portion of the harvested colostrum to ensure they receive the necessary nutrients. After the colostrum is harvested it is rapidly cooled in water before being stored in the freezer at -20°C.
  • Frozen blocks of colostrum are chipped into the raw colostrum tank, using a modified "Butcher Boy" milling machine. Milled colostrum is thawed and recirculated at 7°C and then heated to 30°C, before being diluted by the addition of similar volume of microfiltered water. At this stage, the pH is confirmed to be in the 6.4 - 6.6 range.
  • the diluted colostrum is heated to 55°C before centrifugation for 1 hr, using a Westfalia milk separator with an automatic desludger, to remove fat, somatic cells, cell debris and some bacteria.
  • Colostrum skim is collected into the water-jacketed, pasteurization tank where the product is pasteurized to destroy the pathogenic bacteria.
  • the maximum temperature it is exposed to is 65°C, which ensures that the immunoglobulin remains active.
  • Colostrum is pasteurized at a minimum temperature of 63°C for 30 minutes, cooled to 5°C and then subjected to membrane ultra-filtration, removing much of the water, ions and lactose.
  • the ultra-filtration step using an APV spiral wound ultrafiltration unit with 10kD membranes, results in the production of a finished product with high protein (over 80% protein) and over 35% IgG, the key component of the product.
  • Ultrafiltration produces a concentrate with about half of the initial volume of the colostrum, by removing water and lactose and concentrates the protein content to around 18% and total solids content to approximately 20%. In general, an amount of IgG of not less than 7% is desirable.
  • the colostrum wet concentrate at 4°C is transferred into freeze-drying trays and lyophilized at -20°C to produce a powder, which is milled to 200 microns.
  • the immunization of cows with several strains of enteropathogenic E. coli boosts the concentration of specific antibodies against these microbes in the blood and colostrum.
  • a patented killed vaccine was used to produce very high levels of specific antibodies against selected surface antigens from 13 of the most common strains of E. coli.
  • the resulting antibodies were enriched with anti-LPS antibodies.
  • the specific antibody titers in an anti-LPS immunoglobulin enriched colostrum preparation hyperimmune BC was analyzed by a validated in-house ELISA against a pool of antigens from both the multiple E. coli strain vaccine (06, 08, 015, 025, 027, 063, 0114, 0115, 0128, 0148, 0153 and 0159 serotypes) and the single-strain (078) vaccine antigens.
  • ELISA methods were as follows. First an anti-LPS immunoglobulin enriched colostrum preparation colostrum powders were weighed and resuspended vigorously in PBS-T at 40mg/ml. Suspensions were vortexed repeatedly over 2-3 hrs at room temperature and un-dissolved components were removed by centrifugation. Supernatant was then collected into fresh tubes for further dilution. Diluted samples in 0.5% casein-PBS-T were made by serial four-fold steps, to give dilutions of 1/250, 1/1000, 1/4000 and 1/16,000, prior to assay.
  • coating antigen (described above) was dispensed into a flat-bottomed 96 well plate (Nunc-Immuno, Nunc, Denmark) and then incubated overnight at 4°C.
  • One ELISA plate was coated with single strain antigen at 1 x 10 -3 mg/ml and another plate was coated with multi-strain antigen at 1 x 10 -2 mg/ml.
  • plates were washed five times with PBS-T and then tapped dry on paper towel. 200 ⁇ l of blocking solution (5% casein-PBS) was added and the plates were incubated for 2hr at 37°C to reduce non-specific binding.
  • Figures 54-55 show the results of this assay, which clearly demonstrates the high content of anti LPS antibodies in an anti-LPS immunoglobulin enriched colostrum preparation.
  • TNBS haptinizing agent 2,4,6-trinitrobenzene sulfonic acid
  • TNBS colitis generally is induced by intrarectal application of TNBS in ethanol.
  • DTH delayed-type hypersensitivity
  • the first period is the sensitization phase.
  • the application of the hapten in TNBS results in modification of autologous molecules in the mucosa, leading to priming of antigen-specific T cells.
  • the initial sensitization phase is about 7 days.
  • the animals were sensitized for TNBS using another site, e.g., the skin.
  • the TNBS-induced colitis was scored on day +4, which reflects this acute local reaction representing the initial sensitizing phase.
  • the efferent or elicitation phase, the DTH reaction can be studied only after reapplication of the initiating agents.
  • mice Male, Balb-c, 7-8 weeks old nice were maintained on standard laboratory chow and kept in 12-h light/dark cycles. The mice were moved to an SPF-free environment (laboratory) 3 days before commencing the experiment. To induce hapten-mediated colitis, the mice were sensitized with 160mL of the haptenizing agent TNBS (Sigma-Aldrich, Rhovot, Israel) at a concentration of 2.5% in 50% ethanol by skin painting on day -7. On day 0, 120mL of 2.5% TNBS in 50% ethanol was administered intrarectally via a 3.5-French catheter which was carefully inserted into the colon such that the tip was 4 cm proximal to the anus.
  • TNBS Sigma-Aldrich, Rhovot, Israel
  • Animals were then kept in a vertical position for 30 seconds and returned to their cages. Animals were treated orally (see tables 8 and 9) by various treatments during and after the senstization phase, and then sacrificed on day +4 (four days after the intrarectal TNBS installation.
  • Weight is one of the key parameters in the TNBS-model. In order to assess weight loss, all mice were weighed on day -7 (beginning of experiment), on day 0 (intrarectal TNBS) and on the sacrifice day. The graphs shown in Figure 56 represent weight loss (% of weight loss from their starting weight). The data is shown in Table 10.
  • TNBS-colitis is resulting from mucosal inflammation mediated by a Th1 response
  • many components of the immune system are involved. Changes in the distribution of regulatory T cells in the spleen and liver of treated mice were examined by flow cytometry after sacrificing the animals. Isolation of splenocytes and intrahepatic lymphocytes was performed as follows. Livers and spleens were kept in RPMI-1640 supplemented with FCS. Spleens were crushed through a 70-mm nylon cell strainer 26 and centrifuged (1250 rpm for 7 min). Red blood cells were lysed in 1ml of cold 155mM ammonium chloride lysis buffer.
  • Splenocytes were washed and resuspended in 1ml of RPMI supplemented with FCS. Viability assessed by trypan blue exclusion was above 90%.
  • livers were crushed through a stainless mesh (size 60, Sigma).
  • 10ml of Lymphoprep (Ficoll, Axis-Shield PoC AS, Oslo, Norway) was loaded with a similar volume of cell suspension in 50ml tubes. The tubes were centrifuged at 1800 rpm for 18 min. Cells present in the interface were collected and centrifuged again at 1800 rpm for 10 min to obtain a pellet of cells depleted of hepatocytes. Approximately 1x10 6 cells/mouse liver, were recovered.
  • Flow cytometry for lymphocyte subsets was performed as follows. Flow cytometry was performed following splenocyte and hepatic lymphocyte isolation using 1x10 6 lymphocytes in 100ml PBS with 0.1% BSA. For surface staining, cells were incubated with fluorochrome-conjugated antibodies to the indicated cell-surface markers (eBioscience, San Diego, CA, USA) at the recommended dilution or with isotype control antibodies for 30 minutes at 4°C. The following cell surface anti-mouse antibodies were used: CD4-eFluoro 450, CD8- FITC and CD25-PE. Cells were then washed in PBS containing 1% BSA and fixed with fixation buffer (eBioscience) for another 50 minutes.
  • fluorochrome-conjugated antibodies to the indicated cell-surface markers (eBioscience, San Diego, CA, USA) at the recommended dilution or with isotype control antibodies for 30 minutes at 4°C.
  • the following cell surface anti-mouse antibodies
  • CD4+CD25+ cells were augmented by treatment of colostrum control and 500mg dose of anti LPS, however, the 50mg dose of anti LPS caused a significant decrease in the expression of this marker in the spleen.
  • CD4+FOXP3+ cells were increased by the higher dose of anti LPS, when compared to colostrum control.
  • Serum IL-10 levels were also determined, and the higher dose (500 ug) induced an increase in serum levels of this anti-inflammatory cytokine ( Figure 60 ).
  • Example 49 Anti-LPS Enriched Immunoglobulin Preparation Ameliorates Symptoms of Infection
  • the preparation was tested in a sepsis model.
  • C57Bl/6, 11-12 weeks old (25gr) mice were fed with BCP control or Anti-LPS enriched immunoglobulin colostrum preparation for 10 days.
  • Sepsis was induced by IP injection of 1 mg LPS and 20mg of Gal/N on the last day of the experiment, 5 hours before sacrificing the mice.
  • LAL Limulus amebocyte lysate

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Claims (5)

  1. Composition comprenant une préparation de colostrum enrichie en immunoglobuline anti-LPS destinée à être utilisée dans le traitement d'un sujet atteint d'une stéatohépatite non alcoolique (SHNA), le sujet présentant :
    un niveau ALT supérieur à 50 IU/dl ;
    un niveau AST supérieur à 50 IU/dl ;
    une PA supérieure à 70U/I ; et/ou
    une GGT supérieure à 60 U/I,
    la composition étant formulée pour une administration orale, la forme posologique orale comprenant de 200 mg à 2000 mg d'IgG, et l'immunoglobuline anti-LPS représentant au moins 7 % en poids sec de la composition d'IgG.
  2. Composition destinée à être utilisée selon la revendication 1, dans laquelle la préparation d'immunoglobuline anti-LPS est préparée en immunisant une vache avec des LPS comprenant des LPS 06, 08, 015, 025, 027, 063, 078, 0114, 0115, 0128, 0148, 0153 et 0159 provenant d'une souche d'E.coli entérotoxigénique.
  3. Composition destinée à être utilisée selon l'une quelconque des revendications précédentes, dans laquelle la préparation d'immunoglobuline anti-LPS est administrée à une dose comprise entre 500 mg ou plus à 2000 mg ou plus par jour.
  4. Composition destinée à être utilisée selon l'une quelconque des revendications précédentes, dans laquelle la préparation d'immunoglobuline anti-LPS est administrée à une dose supérieure à 1800 mg par jour.
  5. Composition destinée à être utilisée selon l'une quelconque des revendications précédentes, la composition étant administrée pendant au moins 14 jours.
EP11793860.5A 2010-08-17 2011-08-17 Préparation d'immunoglobuline enrichie en anti-lps, destinée à être utilisée dans le traitement et/ou la prophylaxie d'un trouble pathologique Not-in-force EP2605791B1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US37432810P 2010-08-17 2010-08-17
PCT/IB2011/002596 WO2012023051A2 (fr) 2010-08-17 2011-08-17 Préparation d'immunoglobuline enrichie en anti-lps, destinée à être utilisée dans le traitement et/ou la prophylaxie d'un trouble pathologique

Publications (2)

Publication Number Publication Date
EP2605791A2 EP2605791A2 (fr) 2013-06-26
EP2605791B1 true EP2605791B1 (fr) 2017-03-08

Family

ID=45218767

Family Applications (1)

Application Number Title Priority Date Filing Date
EP11793860.5A Not-in-force EP2605791B1 (fr) 2010-08-17 2011-08-17 Préparation d'immunoglobuline enrichie en anti-lps, destinée à être utilisée dans le traitement et/ou la prophylaxie d'un trouble pathologique

Country Status (8)

Country Link
US (1) US9943597B2 (fr)
EP (1) EP2605791B1 (fr)
AU (1) AU2011290478B2 (fr)
CA (1) CA2808361A1 (fr)
DK (1) DK2605791T3 (fr)
ES (1) ES2628032T3 (fr)
HK (1) HK1185016A1 (fr)
WO (1) WO2012023051A2 (fr)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3461840A1 (fr) 2009-04-27 2019-04-03 Immuron Limited Préparation d'immunoglobuline enrichie en anti-lps destinée au traitement et/ou à la prophylaxie de la stéatohépatite non alcoolique
CA2813612A1 (fr) * 2010-10-04 2012-04-12 Immuron Limited Methodes et compositions utilisant des anticorps anti-ligand lps destinees au traitement et a la prevention de troubles inflammatoires
DE102011006781A1 (de) * 2011-04-05 2012-10-11 Mat-Malta Advanced Technologies Limited Antikörperprodukt, umfassend n spezifische Antikörper
DE102011006809A1 (de) 2011-04-05 2012-10-11 Freistaat Bayern vertreten durch die Julius-Maximilians-Universität Würzburg Verwendung eines Mittels aus Antikörpern und/oder Insulin-like growth factor-Antagonisten
WO2012171077A1 (fr) * 2011-06-17 2012-12-20 Immuron Limited Procédé et composition pour traiter ou inhiber une mucosite associée à une chimiothérapie ou à une lésion causée par un rayonnement
WO2014121045A1 (fr) * 2013-01-31 2014-08-07 The University Of Maryland, Baltimore Composition de colostrum enrichie en anticorps anti-endotoxines
AU2017260881A1 (en) * 2016-05-06 2018-12-20 Hadasit Medical Research Services & Development Limited Hyperimmune colostrum in the modulation and treatment of conditions associated with the mammailan microbiome
CN108095767B (zh) * 2018-01-19 2020-10-02 无锡海斯凯尔医学技术有限公司 组织炎症活动度检测装置
US20210217523A1 (en) * 2018-05-18 2021-07-15 Shimadzu Corporation Diagnosis assistance system and diagnosis assistance device
EP3853356A4 (fr) * 2018-09-20 2022-09-14 Mystem Biotechnologies Inc. Cellules souches dérivées du colostrum, différenciation neuronale, compositions et suppléments pour améliorer la santé des mammifères
WO2021168321A1 (fr) * 2020-02-19 2021-08-26 Anubis Bio Corporation Composition et procédés de traitement d'agents infectieux au moyen d'anticorps spécifiques aux pathogènes
WO2022011436A1 (fr) * 2020-07-17 2022-01-20 Immuron Limited Méthodes de traitement d'une infection à coronavirus avec du colostrum hyperimmun bovin
US11869650B2 (en) * 2021-01-12 2024-01-09 Tandem Diabetes Care, Inc. Remote access for medical device therapy

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH627079A5 (en) 1977-04-15 1981-12-31 Nestle Sa Process for preparing a protein concentrate containing immunological factors of milk origin
US4550019A (en) 1978-03-22 1985-10-29 South Africa Inventions Development Corporation Manufacture and use of fowl egg antibodies
JPS58154513A (ja) 1982-03-09 1983-09-14 Sendai Biseibutsu Kenkyusho 予防及び治療薬
US4748018A (en) 1984-02-07 1988-05-31 Stolle Research & Development Corp. Method of passive immunization of mammals using avian antibody
GB9105292D0 (en) 1991-03-13 1991-04-24 Sandoz Ltd Improvements in or relating to organic compounds
EP0725789B1 (fr) 1993-09-20 2003-03-19 Anadis Ltd. Procede d'extraction d'immunoglobulines du colostrum et emploi de ces dernieres dans des preparations pharmaceutiques
US5397372A (en) 1993-11-30 1995-03-14 At&T Corp. MCVD method of making a low OH fiber preform with a hydrogen-free heat source
ID26730A (id) * 1998-05-27 2001-02-01 Gemma Biotechnology Ltd Induksi protein-protein dan peptida-peptida antibiotik oleh lait/scd14-protein
DE19830607C2 (de) 1998-07-09 2002-08-01 Hte Ag The High Throughput Exp Verfahren zum Nachweis eines Produktes im Abstrom eines katalytischen Materials einer Vielzahl von katalytischen Materialien
KR20100137015A (ko) 2002-03-21 2010-12-29 아나디스 리미티드 불안정한 생활성 물질 및 포유동물 초유를 포함한 조성물, 제조 및 치료 방법
IL165300A0 (en) 2002-05-21 2005-12-18 Anadis Ltd Method of prophylaxis of infection
US9701737B2 (en) 2003-02-19 2017-07-11 Camas, Incorporated Immunogen adherence and method of making and using same
AU2003901008A0 (en) * 2003-03-04 2003-03-20 Anadis Ltd Composition for the treatment and prevention of bacterial infections
TW200616546A (en) 2004-09-29 2006-06-01 Asama Kasei Kk A function composition or food containing a whey protein, an antibody from milk or an antibody
US20090169566A1 (en) 2004-11-22 2009-07-02 Anadis Ltd. Bioactive compositions
JP2010501596A (ja) 2006-08-31 2010-01-21 エー・シー・エヌ・135 493 391・プロプライエタリー・リミテッド・アズ・トラスティー・フォー・コンカ・ユニット・トラスト 抗体含有組成物を用いた非感染性の病状の処置および/または予防
WO2009113065A1 (fr) 2008-03-13 2009-09-17 Hadasit Medical Research Services & Development Ltd. Compositions immunomodulatrices pour le traitement de troubles d’origine immunologique
EP3461840A1 (fr) * 2009-04-27 2019-04-03 Immuron Limited Préparation d'immunoglobuline enrichie en anti-lps destinée au traitement et/ou à la prophylaxie de la stéatohépatite non alcoolique

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
ANONYMOUS: "NAFLD & NASH | World Gastroenterology Organisation", 1 March 2016 (2016-03-01), XP055254038, Retrieved from the Internet <URL:http://www.worldgastroenterology.org/guidelines/global-guidelines/nafld-nash> [retrieved on 20160301] *
ANONYMOUS: "Nonalcoholic Fatty Liver Disease - A Growing Public Health Problem | World Gastroenterology Organisation", 1 March 2016 (2016-03-01), XP055254240, Retrieved from the Internet <URL:http://www.worldgastroenterology.org/publications/e-wgn/e-wgn-expert-point-of-view-articles-collection/nonalcoholic-fatty-liver-disease-a-growing-public-health-problem> [retrieved on 20160301] *
ANONYMOUS: "Nonalcoholic Steatohepatitis National Digestive Diseases Information Clearinghouse", 1 November 2009 (2009-11-01), pages 1 - 6, XP055254037, Retrieved from the Internet <URL:http://www.niddk.nih.gov/health-information/health-topics/liver-disease/nonalcoholic-steatohepatitis/Documents/NASH_508.pdf> [retrieved on 20160301] *
DOCCHECK MEDICAL SERVICES GMBH: "Alanin-Aminotransferase - DocCheck Flexikon", 1 March 2016 (2016-03-01), XP055254254, Retrieved from the Internet <URL:http://flexikon.doccheck.com/de/Alanin-Aminotransferase> [retrieved on 20160301] *
DOCCHECK MEDICAL SERVICES GMBH: "Alkalische Phosphatase - DocCheck Flexikon", 1 March 2016 (2016-03-01), XP055254268, Retrieved from the Internet <URL:http://flexikon.doccheck.com/de/Alkalische_Phosphatase> [retrieved on 20160301] *
DOCCHECK MEDICAL SERVICES GMBH: "Aspartat-Aminotransferase - DocCheck Flexikon", 1 March 2016 (2016-03-01), XP055254259, Retrieved from the Internet <URL:http://flexikon.doccheck.com/de/Aspartat-Aminotransferase> [retrieved on 20160301] *
DOCCHECK MEDICAL SERVICES GMBH: "Gamma-Glutamyltransferase - DocCheck Flexikon", 1 March 2016 (2016-03-01), XP055254267, Retrieved from the Internet <URL:http://flexikon.doccheck.com/de/Gamma-Glutamyltransferase> [retrieved on 20160301] *

Also Published As

Publication number Publication date
AU2011290478B2 (en) 2014-11-13
CA2808361A1 (fr) 2012-02-23
US9943597B2 (en) 2018-04-17
EP2605791A2 (fr) 2013-06-26
US20130224216A1 (en) 2013-08-29
DK2605791T3 (en) 2017-06-19
WO2012023051A9 (fr) 2012-06-14
AU2011290478A1 (en) 2013-02-28
ES2628032T3 (es) 2017-08-01
WO2012023051A3 (fr) 2012-04-26
WO2012023051A2 (fr) 2012-02-23
HK1185016A1 (zh) 2014-02-07

Similar Documents

Publication Publication Date Title
EP2605791B1 (fr) Préparation d&#39;immunoglobuline enrichie en anti-lps, destinée à être utilisée dans le traitement et/ou la prophylaxie d&#39;un trouble pathologique
US20190209681A1 (en) Anti-LPS Enriched Immunoglobulin Preparation for Use in the Treatment and/or Prophylaxis of a Pathologic Disorder
US20130164302A1 (en) Immuno-Modulating Compositions for the Treatment of Immune-Mediated Disorders
KR102037885B1 (ko) 광범위한, 미확인 또는 혼합된 임상적 적용에서 치료용 조성물들 및 방법들
Adar et al. Oral administration of immunoglobulin G-enhanced colostrum alleviates insulin resistance and liver injury and is associated with alterations in natural killer T cells
Feeney et al. The role of immunoglobulins from bovine colostrum and milk in human health promotion
US10464998B2 (en) Anti-LPS enriched immunoglobulin for use in treatment and/or prophylaxis of fibrosis
JP4911894B2 (ja) 関節リウマチを予防するための経口投与用組成物
AU690521B2 (en) Anti-cholesterolemic egg, vaccine and method for production,and use

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20130313

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1185016

Country of ref document: HK

17Q First examination report despatched

Effective date: 20141007

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20160923

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

Ref country code: AT

Ref legal event code: REF

Ref document number: 872928

Country of ref document: AT

Kind code of ref document: T

Effective date: 20170315

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602011035760

Country of ref document: DE

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20170612

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170609

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170608

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2628032

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20170801

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 872928

Country of ref document: AT

Kind code of ref document: T

Effective date: 20170308

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 7

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170608

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170708

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170710

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602011035760

Country of ref document: DE

REG Reference to a national code

Ref country code: HK

Ref legal event code: GR

Ref document number: 1185016

Country of ref document: HK

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20171211

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20170831

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170817

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 8

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170831

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170817

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170817

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20110817

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20190826

Year of fee payment: 9

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170308

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IE

Payment date: 20190805

Year of fee payment: 9

Ref country code: ES

Payment date: 20190919

Year of fee payment: 9

Ref country code: DK

Payment date: 20190822

Year of fee payment: 9

Ref country code: FR

Payment date: 20190822

Year of fee payment: 9

Ref country code: SE

Payment date: 20190822

Year of fee payment: 9

Ref country code: DE

Payment date: 20190822

Year of fee payment: 9

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20190828

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20190829

Year of fee payment: 9

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170308

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602011035760

Country of ref document: DE

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20200831

REG Reference to a national code

Ref country code: SE

Ref legal event code: EUG

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: NL

Ref legal event code: MM

Effective date: 20200901

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20200817

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200831

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200831

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200818

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210302

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200831

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200831

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200817

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200817

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200901

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20220110

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200818